AU2012338733B2 - Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation - Google Patents

Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation Download PDF

Info

Publication number
AU2012338733B2
AU2012338733B2 AU2012338733A AU2012338733A AU2012338733B2 AU 2012338733 B2 AU2012338733 B2 AU 2012338733B2 AU 2012338733 A AU2012338733 A AU 2012338733A AU 2012338733 A AU2012338733 A AU 2012338733A AU 2012338733 B2 AU2012338733 B2 AU 2012338733B2
Authority
AU
Australia
Prior art keywords
adm
antibody
adrenomedullin
scaffold
fragment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2012338733A
Other versions
AU2012338733A1 (en
Inventor
Andreas Bergmann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Adrenomed AG
Original Assignee
Adrenomed AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adrenomed AG filed Critical Adrenomed AG
Publication of AU2012338733A1 publication Critical patent/AU2012338733A1/en
Application granted granted Critical
Publication of AU2012338733B2 publication Critical patent/AU2012338733B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/54F(ab')2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/70Mechanisms involved in disease identification
    • G01N2800/7095Inflammation

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Epidemiology (AREA)
  • Endocrinology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Microbiology (AREA)
  • Emergency Medicine (AREA)
  • Diabetes (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)

Abstract

Patients having a chronic or acute disease or acute condition may be in need for stabilizing the circulation, Subject matter of the present disclosure is an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-lg scaffold for use in therapy of acute disease or condition of a patient for stabilizing the circulation. In a preferred embodiment anti-ADM antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-lg scaffold according to the present disclosure reduces the vasopressor requirement, e.g. catecholamine requirement of said patient. The catecholamine requirement of a patient is an indicator for the condition of the circulation and hemodynamic function of said patient.

Description

PCT/EP2012/072932 WO 2013/072513
Auti-Adrenomcdullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation
Field of the invention
Subject matter of the present invention is an anti-adrenomedullin (ADM) antibody or an anti-adrenomeduliin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or condition of a patient for stabilizing the circulation.
Background
The peptide adrenomedullin (ADM) was described for the first time in 1993 (Kitamura, K., et ah, "Adrenomedullin: A Novel Hypotensive Peptide Isolated From Human Pheochromocytoma", Biochemical and Biophysical Research Communications, Vol. 192 (2), pp. 553-560 (1993)) as a novel hypotensive peptide comprising 52 amino acids, which had been isolated from a human pheochromocytome; SEQ ID No.: 21. In the same year, cDNA coding for a precursor peptide comprising 185 amino acids and the complete amino acid sequence of this precursor peptide were also described. The precursor peptide, which comprises, inter alia, a signal sequence of 21 amino acids at the N-terminus, is referred to as "preproadrenomedullin" (pre-proADM). In the present description, all amino acid positions specified usually relate to the pre-proADM which comprises the 185 amino acids. The peptide adrenomedullin (ADM) is a peptide which comprises 52 amino acids (SEQ ID NO: 21) and which comprises the amino acids 95 to 146 of pre-proADM, from which it is formed by proteolytic cleavage. To date, substantially only a few fragments of the peptide fragments formed in the cleavage of the pre-proADM have been more exactly characterized, in particular the physiologically active peptides adrenomedullin (ADM) and "PAMP", a peptide comprising 20 amino acids (22-41) which follows the 21 amino acids of the signal peptide in pre-proADM. The discovery and characterization of ADM in 1993 triggered intensive research activity, the results of which have been summarized in various review articles, in the context of the present description, reference being made in particular to the articles to be found in an issue of "Peptides" devoted to ADM in particular (Editorial, Takahashi, K., "Adrenomedullin: from a pheochromocytoma to the eyes", Peptides, Vol. 22, p. 1691 (2001)) and (Eto, T., "A review of the biological properties and clinical implications of WO 2013/072513 PCT/EP2012/072932 2 adrenomedullin and proadrenomedullin N-terminal 20 peptide (PAMP), hypotensive and vasodilating peptides", Peptides, Vol. 22, pp, 1693-1711 (2001)). A further review is (Hinson, et al, "Adrenomedullin, a Multifunctional Regulatory Peptide", Endocrine Reviews, Vol. 21(2), pp. 138-167 (2000)). In the scientific investigations to date, it has been found, inter alia, that ADM may be regarded as a polyfimctional regulatory peptide. It is released into the circulation in an inactive form extended by glycine (Kitamura, K., et al, "The intermediate form of glycine-extended adrenomedullin is the major circulating molecular form in human plasma", Biochem. Biophys. Res. Commun., Vol. 244(2), pp. 551-555 (1998). Abstract Only). There is also a binding protein (Pio, R., et al„ "Complement Factor H is a Serum-binding Protein for adrenomedullin, and the Resulting Complex Modulates the Bioactivities of Both Partners", The Journal of Biological Chemistry, Vol. 276(15), pp. 12292-12300 (2001)) which is specific for ADM and probably likewise modulates the effect of ADM. Those physiological effects of ADM as well as of PAMP which are of primary importance in the investigations to date were the effects influencing blood pressure.
Hence, ADM is an effective vasodilator, and it is possible to associate the hypotensive effect with the particular peptide segments in the C-terminal part of ADM. It has furthermore been found that the above-mentioned further physiologically active peptide PAMP formed from pre-pro ADM likewise exhibits a hypotensive effect, even if it appears to have an action mechanism differing from that of ADM (cf. in addition to the abovementioned review articles (Eto, T., "A review of the biological properties and clinical implications of adrenomedullin and proadrenomedullin N-terminal 20 peptide (PAMP), hypotensive and vasodilating peptides", Peptides, Vol. 22, pp. 1693-1711 (2001)) and (Hinson, et al, "Adrenomedullin, a Multifunctional Regulatory Peptide", Endocrine Reviews, Vol. 21(2), pp. 138-167 (2000)) also (Kuwasako, K., et al, "Purification and characterization of PAMP-12 (PAMP-20) in porcine adrenal medulla as a major endogenous biologically active peptide", FEBS Lett, Vol. 414(1), pp. 105-110 (1997). Abstract only), (Kuwasaki, K., et al, "Increased plasma proadrenomedullin N-terminal 20 peptide in patients with essential hypertension", Ann. Clin. Biochem., Vol. 36 (Pt. 5), pp. 622-628 (1999). Abstract only) or (Tsuruda, T., et al, "Secretion of proadrenomedullin N-terminal20 peptide from cultured neonatal rat cardiac cells", Life Sci., Vol. 69(2), pp. 239-245 (2001). Abstract only) and EP-A2 0 622 458). It has furthermore been found that the concentrations of ADM which can be measured in the circulation and other biological liquids are, in a number of pathological states, significantly above the concentrations to be found in healthy control persons. Thus, the ADM level in WO 2013/072513 PCT/EP2012/072932 3 patients with congestive heart failure, myocardial infarction, kidney diseases, hypertensive disorders, Diabetes mellitus, in the acute phase of shock and in sepsis and septic shock are significantly increased, although to different extents. The PAMP concentrations are also increased in some of said pathological states, but the plasma levels are lower relative to ADM ((Eto, T., "A review of the biological properties and clinical implications of adrenomedullin and pro adrenomedullin N-terminal 20 peptide (PAMP), hypotensive and vasodilating peptides", Peptides, Vol. 22, pp. 1693-1711 (2001)); page 1702). It is furthermore known that unusually high concentrations of ADM are to be observed in sepsis, and the highest concentrations in septic shock (cf. (Eto, T., "A review of the biological properties and clinical implications of adrenomedullin and proadrenomedullin N-terminal 20 peptide (PAMP), hypotensive and vasodilating peptides", Peptides, Vol. 22, pp. 1693-1711 (2001)) and (Hirata, et al, "Increased Circulating Adrenomedullin, a Novel Vasodilatory Peptide, in Sepsis", Journal of Clinical Endocrinology and Metabolism, Vol. 81(4), pp. 1449-1453 (1996)), (Ehlenz, K., et al, "High levels of circulating adrenomedullin in severe illness: Correlation with C-reactive protein and evidence against the adrenal medulla as site of origin", Exp Clin Endocrinol Diabetes, Vol. 105, pp. 156-162 (1997)), (Tomoda, Y., et al., "Regulation of adrenomedullin secretion from cultured cells", Peptides, Vol. 22, pp. 1783-1794 (2001)), (Ueda, S., et al, "Increased Plasma Levels of Adrenomedullin in Patients with Systemic Inflammatory Response Syndrome", Am. J. Respir. Crit. Care Med., Vol. 160, pp. 132-136 (1999)) and (Wang, P., "Adrenomedullin and cardiovascular responses in sepsis", Peptides, Vol. 22, pp. 1835-1840 (2001)).
Known in the art is further a method for identifying adrenomedullin immunoreactivity in biological liquids for diagnostic purposes and, in particular within the scope of sepsis diagnosis, cardiac diagnosis and cancer diagnosis. According to the invention, the midregional partial peptide of the proadrenomedullin, which contains amino acids (45-92) of the entire preproadrenomedullin, is measured, in particular, with an immunoassay which works with at least one labeled antibody that specifically recognizes a sequence of the mid-proADM (W02004/090546). WO-A1 2004/097423 describes the use of an antibody against adrenomedullin for diagnosis, prognosis, and treatment of cardiovascular disorders. Treatment of diseases by blocking the ADM receptor are also described in the art, (e.g. WO-A1 2006/027147, PCT/EP2005/012844) said diseases may be sepsis, septic shock, cardiovascular diseases, infections, dermatological WO 2013/072513 PCT/EP2012/072932 4 diseases, endocrinological diseases, metabolic diseases, gastroenterological diseases, cancer, inflammation, hematological diseases, respiratory diseases, muscle skeleton diseases, neurological diseases, urological diseases.
It is reported for the early phase of sepsis that ADM improves heart function and the blood supply in liver, spleen, kidney and small intestine. ADM-neutralizing antibodies neutralize the before mentioned effects during the early phase of sepsis (Wang, P., "Adrenomedullin and cardiovascular responses in sepsis”, Peptides, Vol. 22, pp. 1835-1840 (2001).
In the later phase of sepsis, the hypodynamical phase of sepsis, ADM constitutes a risk factor that is strongly associated with the mortality of patients in septic shock. (Schiitz et al., “Circulating Precursor levels of endothelin-1 and adrenomedullin, two endothelium-derived, counteracting substances, in sepsis”, Endothelium, 14:345-351, (2007)). Methods for the diagnosis and treatment of critically ill patients, e.g. in the very late phases of sepsis, and the use of endothelin and endothelin agonists with vasoconstrictor activity for the preparation of medicaments for the treatment of critically ill patients have been described in WO-A1 2007/062676. It is further described in WO-A1 2007/062676 to use, in place of endothelin and/or endothelin agonists, or in combination therewith, adrenomedullin antagonists, i.e. molecules which prevent or attenuate the vasodilating action of adrenomedullin, e.g. by blocking its relevant receptors, or substances preventing the binding of adrenomedullin to its receptor (e.g. specific binders as e.g. antibodies binding to adrenomedullin and blocking its receptor bindings sites; “immunological neutralization44). Such use, or combined use, including a subsequent or preceding separate use, has been described in certain cases to be desirable for example to improve the therapeutic success, or to avoid undesirable physiological stress or side effects. Thus, it is reported that neutralizing ADM antibodies may be used for the treatment of sepsis in the late stage of sepsis.
Administration of ADM in combination with ADM-binding-Protein-1 is described for treatment of sepsis and septic shock in the art. It is assumed that treatment of septic animals with ADM and ADM-binding-Protein-l prevents transition to the late phase of sepsis. It has to be noted that in a living organism ADM binding protein (complement factor H) is present in the circulation of said organism in high concentrations (Pio et al.: Identification, characterization, and physiological actions of factor H as an Adrenomedullin binding Protein present in Human Plasma; Microscopy Res. and Technique, 55:23-27 (2002) and Martinez et H:\grs\Intcrwovcn\NRPorihl\DCC\GRS\l5l7478l_l.docx-l 1/07/2017 2012338733 11 Μ 2017 5 al.\ Mapping of the Adrenomedullin-Binding domains in Human Complement factor H; Hypertens Res Vol. 26, Suppl (2003), S56-59).
In accordance with the invention the ADM-binding-Protein-1 may be also referred to as 5 ADM-binding-Protein-1 (complement factor H).
Patients having a chronic or acute disease or acute condition may be in need for stabilizing the circulation or their hemodynamic function (Cavazonni and Dellinger, Critical Care 2006, 10(Suppl 3):S2 (doi:10.1186/cc4829). 10
According to a first aspect of the present invention there is provided a method of stabilizing the systemic circulation of a patient in need thereof by preventing or treating an acute disease or acute condition in said patient, the method comprising administering an effective dose of an anti-Adrenomedullin (ADM) antibody, an anti-ADM antibody fragment binding to 15 adrenomedullin or an anti-ADM non-Ig protein scaffold binding to adrenomedullin to said patient, wherein said antibody, antibody fragment or non-IG scaffold binds to a region of at least 4 amino acids within the sequence of amino acids 1-42 of mature human ADM as set out in SEQ ID NO: 24. 20 According to a second aspect of the present invention there is provided the use of an anti-Adrenomedullin (ADM) antibody, anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig protein scaffold binding to adrenomedullin in the manufacture of a medicament for preventing or treating an acute disease or acute condition in a patient in need thereof, wherein said antibody, antibody fragment or non-Ig scaffold binds to a region of at 25 least 4 amino acids within the sequence of amino acids 1 -42 of mature human ADM as set out in SEQ ID NO: 24.
Description of the invention
Subject matter of the present invention is an anti-adrenomedullin (ADM) antibody or an anti-30 adrenomedullin antibody fragment binding to ADM or anti-ADM non-Ig scaffold binding to ADM for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation, in particular the systemic circulation of said patient. In particular, subj ect matter of 2012338733 11 Μ 2017 M:\grs\InicrwOvcn\NRPorihl\DCC\GRS\l5l7478l_l docx-l 1/07/2017 5a the present invention is an anti-adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment binding to ADM or anti-ADM non-Ig scaffold binding to ADM for use in therapy of an acute disease or acute condition of a patient for stabilizing the systemic circulation of said patient wherein said patient is in need of stabilizing the circulation. 5
Systemic circulation refers to the part of the circulatory system in which the blood leaves the heart, services the body's cells, and then re-enters the heart. Blood leaves through the left ventricle to the aorta, the body's largest artery. The aorta leads to smaller arteries, arterioles, and finally capillaries. Waste and carbon dioxide diffuse out of the cell into the blood, and 10 oxygen in the blood diffuses into the cell. Blood then moves to venous capillaries, and then to the venae cavae: the lower inferior vena cava and the upper superior vena cava, through which the blood re-enters the heart at the right atrium.
Throughout the specification stabilizing the circulation means stabilizing the systemic 15 circulation. The term systemic circulation would not encompass phenomena of microcirculation. Microcirculation is the delivery of fresh blood to the smallest blood vessels, present in the vasculature embedded within organ tissues. This contrasts with macrocirculation, which transport blood to and from the organs. The state of the systemic circulation may be measures by parameters like mean arterial pressure, blood pressure (other WO 2013/072513 PCT/EP2012/072932 6 parameters see above). A patient in need for stabilizing the circulation may be, thus a patient that exhibits a heart rate of > 100 beats /min and or < 65 mm Hg mean arterial pressure. If the circulation is stabilized by the administration of an anti-Adrenomeduilin (ADM) antibody or by an anti-ADM antibody fragment binding to adrenomedullin or an anti-ADM non-Ig scaffold binding to adrenomedullin, this can be measured and is characterized by an increase of the mean arterial pressure over 65 mm Hg and/or an decrease of heart rate under 100 beats/min.
It should be emphasized that the provided anti-adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold are intended by the present invention to be applied for sake of stabilizing the systemic circulation, and thus are not necessarily intended for any methods of primary treatment or first line treatment to the acute disease or acute condition itself that has to be considered as underlying disease(s). This means the present invention do not provide for a therapy of healing/curing e.g. cancer, diabetes, meningitis, polytrauma, and the like. Accordingly, the therapy for an acute disease or acute condition of a patient within the scope of the invention is related to any kind of systemic circulatory insufficiency, or poor systemic circulation of the blood as an acute event.
Acute disease or acute conditions may be selected from the group but are not limited to the group comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), or sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy. Especially useful is the antibody or fragment or scaffold according to the present invention for reducing the risk of mortality during sepsis and septic shock, i.e. late phases of sepsis.
In the following clinical criteria for SIRS, sepsis, severe sepsis, septic shock will be defined. 1) Systemic inflammatory host response (SIRS) characterized by at least two of the following symptoms • patients exhibit hypotension (mean arterial pressure is < 65 mm Hg)
• elevated serum lactate level being > 4 mmol/L • blood glucose > 7.7 mmol/L (in absence of diabetes) • central venous pressure is not within the range 8-12 mm Hg • urine output is < 0.5 mL x kg"1 x hr"1 7 • central venous (superior vena cava) oxygen saturation is < 70% or mixed venous < 65% • heart rate is > 90 beats/min
• temperature < 36°C or > 38°C • respiratory rate > 20/min • white cell count < 4 or > 12x109/L (leucocytes); > 10% immature neutrophils 2) Sepsis
Following at least two of the symptoms mentioned under 1), and additionally a clinical suspicion of new infection, being: ® cough/sputum/chest pain • abdominal pain/distension/diarrhoea ® line infection ® endocarditis • dysuria • headache with neck stiffness • cellulitis/wound/joint infection • positive microbiology for any infection 3) Severe sepsis
Provided that sepsis is manifested in patient, and additionally a clinical suspicion of any organ dysfunction, being:
• blood pressure systolic < 90/mean; < 65mmHG
• lactate > 2 mmol/L
• Bilirubine > 34pmol/L urine output <0.5 mL/kg/h for 2h WO 2013/072513 PCT/EP2012/072932 • WO 2013/072513 PCT/EP2012/072932 8
• creatinine >177 μιηοΙ/L
• platelets < 100xl09/L • Sp02 > 90% unless 02 given 4) Septic shock
At least one sign of end-organ dysfunction as mentioned under 3) is manifested. Septic shock is indicated, if there is refractory hypotension that does not respond to treatment and intravenous fluid administration alone is insufficient to maintain a patient's blood pressure from becoming hypotensive also provides for an administration of an anti-ADM antibody or an anti-ADM antibody fragment or an anti-ADM non-Ig scaffold in accordance with the present invention.
In one embodiment of the present invention the patient is not suffering from SIRS, a severe infection, sepsis, shock as e.g. septic shock. Said severe infection denotes e.g. meningitis, Systemic inflammatory Response-Syndrome (SIRS), sepsis, severe sepsis, and shock as e.g. septic shock. In this regard, a severe sepsis is characterized in that sepsis is manifested in said patient, and additionally a clinical suspicion of any organ dysfunction is present, being it:
• blood pressure systolic < 90/mean; < 65mmHG
• lactate > 2 mmol/L
• Bilirubine > 34pmol/L • urine output <0.5 mL/kg/h for 2h
• creatinine >177 pmol/L
• platelets < 1 OOx 109/L • Sp02 > 90% unless 02 given
In another embodiment said acute disease or acute condition is not sepsis, or not severe sepsis, or not SIRS, or not shock, or not septic shock. WO 2013/072513 PCT/EP2012/072932 9
In another embodiment said acute disease or acute condition is not sepsis.
In another embodiment said acute disease or acute condition is selected from the group selected meningitis, diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy.
In all of the above mentioned acute diseases and conditions there might be the need for stabilizing the circulation of a patient by the administration of an anti-adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold, an anti-adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold may also be administered preventively in patients having an acute disease or condition in order to prevent that the heart rate increases of > 100 beats /min and/ or mean arterial pressure decreases to < 65 mm Hg.
Anti-Adrenomedullin (ADM) antibody is an antibody that binds specifically to ADM, Anti-adrenomedullin antibody fragment is a fragment of an anti-ADM antibody, wherein said fragment binds specifically to ADM. An anti-ADM non-Ig scaffold is a non-Ig scaffold that binds specifically to ADM. Specifically binding to ADM allows binding to other antigens as well. This means, this specificity would not exclude that the antibody may cross-react with other polypeptides than that against it has been raised.
In one embodiment the anti-ADM antibody or the anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention reduces the vasopressor-agents requirement, e.g. catecholamine requirement, of said patient. The vasopressor-agents requirement, e.g catecholamine requirement of a patient is an indicator for the condition of the circulation of said patient. Thus, the anti-ADM antibody or the anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold may be administered at a point of time when the patient is in need of a vasopressor agent, e.g. catecholamine.
In one embodiment of the invention said patient is a patient in need of increasing the blood pressure. A patient in need of stabilizing the circulation may a patient with low cardiac output and /or a low blood pressure (hypotension). This may be a patient with a heart rate >100 beats/min. WO 2013/072513 PCT/EP2012/072932 10
This may be a patient with mean arterial pressures < 65 mmHg or even with < 60 mmHg. Mean arterial pressure is defined as MAP = (COxSVR)+CVP where CO is cardiac output; SVR is systemic vascular resistance and CVP is central venous pressure and usually small enough to be neglected in this formula. A patient in need of stabilizing the circulation may be also a patient having in addition to the above symptoms a respiratory rate > 20 /min.
In a specific embodiment of the invention a patient in need of stabilizing the circulation may be a patient with low cardiac output and /or a low blood pressure (hypotension). This may be a patient with a heart rate > 90 beats/min. This may be a patient with mean arterial pressures < 65 mmHg or even with < 60 mmHg.
Some patients with sepsis-induced hypofusion may remain hypotensive despite adequate fluid replacement. In these cases vasopressor agents are needed to increase MAP. Thus, in one embodiment of the invention the patient having a chronic or acute disease or acute condition is a patient in need of vasopressor agents to increase MAP. Catecholamines such as dopamine, epinephrine (adrenaline), norepinephrine (noradrenaline), and phenylephrine have been traditionally used to raise blood pressure in patients with e.g. septic shock. Recently also vasopressin has been suggested as potential vasopressor in patients with a chronic or acute disease or acute condition in need for stabilizing the circulation.
Vasopressor agents as catecholamine may stabilize the circulation of a patient having a chronic or acute disease or acute condition. In case the condition of the patient (low blood pressure) is very critical, vasopressor agents administration, e.g. catecholamine administration, alone may not prevent the break-down of the circulation. The additional administration of anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold together with administration of e.g. catecholamine may help to stabilize the circulation of a patient whose condition is so critical that catecholamine administration without administration of anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold would not be sufficient in order to stabilize the circulation of said patient.
Further, vasopressors may have serious side effects. Dopamine stimulates D1 receptors in the renal regional circulation, producing vasodilation and increases blood flow. This is one of the reasons why clinicians have utilized low doses of dopamine to protect kidney function. Also for other vasopressors it has been suggested that increasing the blood pressure with certain WO 2013/072513 PCT/EP2012/072932 11 drugs, despite its intuitive appeal as something beneficial, can be associated with worse outcomes.
Thus, subject of the invention is an anti-adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient in order to replace the administration of a vasopressor totally or partially. This means the patient according to the present invention may be a patient being in need of or treatment with vasopressors or a patient receiving a treatment with vasopressors.
The anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold may be also administered preventively before the patient exhibits any signs of serious circulation problems. This might be the case if the patient has a chronic or acute disease or acute condition where circulation problems may be expected, comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), or sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, damages induced by chemotherapy. Especially useful is the antibody or fragment or scaffold according to the present invention for reducing the risk of mortality during sepsis and septic shock, i.e. late phases of sepsis. The person skilled in the art is aware that said reducing the risk of mortality is associated with the stabilization of the circulation in accordance with the invention. Acute disease or acute condition may be a disease or condition wherein the patient is characterized as being in need of stabilizing the circulation. The need of stabilizing the circulation is characterized as outlined above, namely this may be a patient preferably with a heart rate of > 90 beats/min or even with a heart rate of > 100 beats/min. This may be a patient with mean arterial pressures < 65 mmHg or even with < 60 mmHg. Mean arterial pressure is defined as MAP = (COxSVR)+CVP where CO is cardiac output; SVR is systemic vascular resistance and CVP is central venous pressure and usually small enough to be neglected in this fonnula. A patient in need of stabilizing the circulation may be also a patient having a respiratory rate > 20 /min.
The circulation stabilizing effect of the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold is thus supporting the primary therapy of said chronic WO 2013/072513 PCT/EP2012/072932 12 or acute disease or acute condition. This means in one embodiment that the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold is administered in addition to a first line treatment (primary therapy). In case of a chronic or acute disease or acute condition like a severe infection, SIRS, sepsis or the like the primary therapy would be e.g. the administration of antibiotics. The anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold would stabilize the circulation and would help to prevent worsening of the critical condition of said patient until the e.g. antibiotic administration takes effect. As before mentioned the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold may be administered in a preventive way or in a therapeutic way, this means in order to prevent circulation problems or in order to stabilize the circulation when circulation problems are present in a said patient.
It should be emphasized that the circulation problems comprised by the present invention may be acute circulation problems according to a specific embodiment of the invention.
In one embodiment of the invention an anti-Adrenomedullin (ADM) antibody or an anti-ADM antibody fragment or anti-ADM non-Ig scaffold is to be used in combination with vasopressors e.g. catecholamine wherein said combination is for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation of said patient.
In one embodiment of the invention said patient having a chronic or acute disease or condition being in need for stabilizing the circulation is characterized by the need of said patient to get administration of vasopressors e.g. catecholamine administration.
Subject matter of the invention in one specific embodiment is, thus, an anti-adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment binding to ADM or anti-ADM non-Ig scaffold binding to ADM for use in therapy of a patient in need of an administration of vasopressors, e.g. a catecholamine administration.
Furthermore, in one embodiment of the invention an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold is to be used in combination with fluids administered intravenously, wherein said combination is for use in therapy of a patient having a chronic or acute disease or acute condition of a patient for stabilizing the circulation of said patient. In one embodiment of the invention said patient having a chronic or acute disease or condition being in need for stabilizing the circulation is characterized by the need of said patient to get intravenous fluids. WO 2013/072513 PCT/EP2012/072932 13
In accordance with the invention the need of a patient to get intravenous fluids is also an acute need due to an acute disease or acute condition. This, however, does not exclude an underlying chronic or acute disease the patient is having and that is maybe associated with the acute need for fluids as well as acute need for stabilizing the circulation.
Subject matter of the invention in one specific embodiment is, thus, an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of a patient in need of intravenous fluids.
Furthermore, in one embodiment of the invention an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold is monospecific. Monospecific anti-Adrenomedullin (ADM) antibody or monospecific anti-adrenomedullin antibody fragment or monospecific anti-ADM non-Ig scaffold means that said antibody or antibody fragment or non-Ig scaffold binds to one specific region encompassing at least 5 amino acids within the target ADM. Monospecific anti-Adrenomedullin (ADM) antibody or monospecific anti-adrenomedullin antibody fragment or monospecific anti-ADM non-Ig scaffold are anti-Adrenomedullin (ADM) antibodies or anti-adrenomedullin antibody fragments or anti-ADM non-Ig scaffolds that all have affinity for the same antigen.
In a specific and preferred embodiment the present invention provides for a monospecific anti-Adrenomedullin (ADM) antibody or monospecific anti-adrenomedullin antibody fragment or monospecific anti-ADM non-Ig scaffold, characterized in that said antibody or antibody fragment or non-Ig scaffold binds to one specific region encompassing at least 4 amino acids within the target ADM.
In another special embodiment the anti-ADM antibody or the antibody fragment binding to ADM is a monospecific antibody. Monospecific means that said antibody or antibody fragment binds to one specific region encompassing preferably at least 4, or at least 5 amino acids within the target ADM. Monospecific antibodies or fragments are antibodies or fragments that all have affinity for the same antigen. Monoclonal antibodies are monospecific, but monospecific antibodies may also be produced by other means than producing them from a common germ cell.
An antibody according to the present invention is a protein including one or more polypeptides substantially encoded by immunoglobulin genes that specifically binds an WO 2013/072513 PCT/EP2012/072932 14 antigen, The recognized immunoglobulin genes include the kappa, lambda, alpha (IgA), gamma (IgGj, IgG2, IgG3, IgG4), delta (IgD), epsilon (IgE) and mu (IgM) constant region genes, as well as the myriad immunoglobulin variable region genes. Full-length immunoglobulin light chains are generally about 25 Kd or 214 amino acids in length. Full-length immunoglobulin heavy chains are generally about 50 Kd or 446 amino acid in length. Light chains are encoded by a variable region gene at the NH2-terminus (about 110 amino acids in length) and a kappa or lambda constant region gene at the COOH-terminus. Heavy chains are similarly encoded by a variable region gene (about 116 amino acids in length) mid one of the other constant region genes.
The basic structural unit of an antibody is generally a tetramer that consists of two identical pairs of immunoglobulin chains, each pair having one light and one heavy chain. In each pair, the light and heavy chain variable regions bind to an antigen, and the constant regions mediate effector functions. Immunoglobulins also exist in a variety of other forms including, for example, Fv, Fab, and (Fab')2, as well as bifunctional hybrid antibodies and single chains (e.g., Lanzavecchia et al., Eur. J. Immunol. 17:105,1987; Huston et al, Proc. Natl. Acad. Sci. US.A., 85:5879-5883, 1988; Bird et al, Science 242:423-426, 1988; Hood et al., Immunology, Benjamin, N.Y., 2nd ed., 1984; Hunkapiller and Hood, Nature 323:15-16,1986). An immunoglobulin light or heavy chain variable region includes a framework region interrupted by three hypervariable regions, also called complementarity determining regions (CDR's) (see, Sequences of Proteins of Immunological Interest, E. Kabat et al, U.S. Department of Health and Human Services, 1983). As noted above, the CDRs are primarily responsible for binding to an epitope of an antigen. An immune complex is an antibody, such as a monoclonal antibody, chimeric antibody, humanized antibody or human antibody, or functional antibody fragment, specifically bound to the antigen.
Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin variable and constant region genes belonging to different species. For example, the variable segments of the genes from a mouse monoclonal antibody can be joined to human constant segments, such as kappa and gamma 1 or gamma 3. In one example, a therapeutic chimeric antibody is thus a hybrid protein composed of the variable or antigen-binding domain from a mouse antibody and the constant or effector domain from a human antibody, although other mammalian species can be used, or the variable region can be produced by molecular techniques. Methods of making chimeric WO 2013/072513 PCT/EP2012/072932 15 antibodies are well known in the art, e.g., see U.S. Patent No. 5,807,715. A "humanized" immunoglobulin is an immunoglobulin including a human framework region and one or more CDRs from a non-human (such as a mouse, rat, or synthetic) immunoglobulin. The nonhuman immunoglobulin providing the CDRs is termed a "donor" and the human immunoglobulin providing the framework is termed an "acceptor." In one embodiment, all the CDRs are from the donor immunoglobulin in a humanized immunoglobulin. Constant regions need not be present, but if they are, they must be substantially identical to human immunoglobulin constant regions, i.e., at least about 85-90%, such as about 95% or more identical. Hence, all parts of a humanized immunoglobulin, except possibly the CDRs, are substantially identical to corresponding parts of natural human immunoglobulin sequences. A "humanized antibody" is an antibody comprising a humanized light chain and a humanized heavy chain immunoglobulin. A humanized antibody binds to the same antigen as the donor antibody that provides the CDRs. The acceptor framework of a humanized immunoglobulin or antibody may have a limited number of substitutions by amino acids taken from the donor framework. Humanized or other monoclonal antibodies can have additional conservative amino acid substitutions which have substantially no effect on antigen binding or other immunoglobulin functions. Exemplary conservative substitutions are those such as gly, ala; val, ile, leu; asp, glu; asn, gin; ser, thr; lys, arg; and phe, tyr. Humanized immunoglobulins can be constructed by means of genetic engineering (e.g., see U.S. Patent No. 5,585,089). A human antibody is an antibody wherein the light and heavy chain genes are of human origin. Human antibodies can be generated using methods known in the art. Human antibodies can be produced by immortalizing a human B cell secreting the antibody of interest. Immortalization can be accomplished, for example, by EBV infection or by fusing a human B cell with a myeloma or hybridoma cell to produce a trioma cell. Human antibodies can also be produced by phage display methods (see, e.g., Dower et al, PCT Publication No. W091/17271; McCafferty et al, PCT Publication No. W092/001047; and Winter, PCT Publication No. WO92/20791), or selected from a human combinatorial monoclonal antibody library (see the Morphosys website). Human antibodies can also be prepared by using transgenic animals carrying a human immunoglobulin gene (for example, see Lonberg et al, PCT Publication No. W093/12227; and Kucherlapati, PCT Publication No. WO91/10741).
Thus, the anti-ADM antibody may have the formats known in the art. Examples are human antibodies, monoclonal antibodies, humanized antibodies, chimeric antibodies, CDR-grafted antibodies. In a preferred embodiment antibodies according to the present invention are WO 2013/072513 PCT/EP2012/072932 16 recombinantly produced antibodies as e.g. IgG, a typical full-length immunoglobulin, or antibody fragments containing at least the F-variable domain of heavy and/or light chain as e.g. chemically coupled antibodies (fragment antigen binding) including but not limited to Fab-fragments including Fab minibodies, single chain Fab antibody, monovalent Fab 5 antibody with epitope tags, e.g. Fab-V5Sx2; bivalent Fab (mini-antibody) dimerized with the CH3 domain; bivalent Fab or multivalent Fab, e.g. formed via multimerization with the aid of a heterologous domain, e.g. via dimerization of dHLX domains,e.g. Fab-dHLX-FSx2; F(abi)2-fragments, scFv-fragments, multimerized multivalent or/and multispecific scFv-fragments, bivalent and/or bispecific diabodies, BITE® (bispecific T-cell engager), 10 trifunctional antibodies, polyvalent antibodies, e.g. from a different class than G; singledomain antibodies, e.g. nanobodies derived from camelid or fish immunoglobulinesand numerous others.
In addition to anti-ADM antibodies other biopolymer scaffolds are well known in the art to 15 complex a target molecule and have been used for the generation of highly target specific biopolymers. Examples are aptamers, spiegelmers, anticalins and conotoxins. For illustration of antibody formats please see Fig. la, lb and lc.
An antibody fragment according to the present invention is an antigen binding fragment of an 20 antibody according to the present invention.
In a preferred embodiment the ADM antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragmentand scFv-Fc Fusion protein. In another preferred embodiment the antibody format is selected from the 25 group comprising scFab fragment, Fab fragment, scFv fragment and bioavailability optimized conjugates thereof, such as PEGylated fragments. One of the most preferred formats is the scFab format.
Non-Ig scaffolds may be protein scaffolds and may be used as antibody mimics as they are 30 capable to bind to ligands or antigenes. Non-Ig scaffolds may be selected from the group comprising tetranectin-based non-Ig scaffolds (e.g. described in US 2010/0028995), fibronectin scaffolds (e.g. described in EP 1266 025; lipocalin-based scaffolds ((e.g, described in WO 2011/154420); ubiquitin scaffolds (e.g. described in WO 2011/073214), transferring scaffolds (e.g. described in US 2004/0023334), protein A scaffolds (e.g. described in EP WO 2013/072513 PCT/EP2012/072932 17 2231860), ankyrin repeat based scaffolds (e.g. described in WO 2010/060748), microproteins preferably microproteins forming a cystine knot) scaffolds (e.g. described in EP 2314308), Fyn SH3 domain based scaffolds (e.g. described in WO 2011/023685) EGFR-A-domain based scaffolds (e.g. described in WO 2005/040229) and Kunitz domain based scaffolds (e.g. described in EP 1941867).
In one embodiment of the invention antibodies according to the present invention may be produced as follows: A Balb/c mouse was immunized with 100 pg ADM-Peptide-BSA-Conjugate at day 0 and 14 (emulsified in 100μ1 complete Freund’s adjuvant) and 50pg at day 21 and 28 (in 100μ1 incomplete Freund’s adjuvant). Three days before the fusion experiment was performed, the animal received 50pg of the conjugate dissolved in 100μ1 saline, given as one intraperitoneal and one intravenous injection.
Spenocytes from the immunized mouse and cells of the myeloma cell line SP2/0 were fused with 1ml 50% polyethylene glycol for 30s at 37°C. After washing, the cells were seeded in 96-well cell culture plates. Hybrid clones were selected by growing in HAT medium [RPMI 1640 culture medium supplemented with 20% fetal calf serum and HAT-Supplement]. After two weeks the HAT medium is replaced with HT Medium for three passages followed by returning to the normal cell culture medium.
The cell culture supernatants were primary screened for antigen specific IgG antibodies three weeks after fusion. The positive tested microcultures were transferred into 24-well plates for propagation. After retesting, the selected cultures were cloned and recloned using the limiting-dilution technique and the isotypes were determined (see also Lane, R.D. (1985). A short-duration polyethylene glycol fusion technique for increasing production of monoclonal antibody-secreting hybridomas. J. Immunol. Meth. 81: 223-228; Ziegler, B. et a/.(1996) Glutamate decarboxylase (GAD) is not detectable on the surface of rat islet cells examined by cytofluorometry and complement-dependent antibody-mediated cytotoxicity of monoclonal GAD antibodies, Horm. Metab. Res. 28: 11-15).
Antibodies may be produced by means of phage display according to the following procedure: WO 2013/072513 PCT/EP2012/072932 18
The human naive antibody gene libraries HAL7/8 were used for the isolation of recombinant single chain F-Variable domains (scFv) against adrenomedullin peptide. The antibody gene libraries were screened with a panning strategy comprising the use of peptides containing a biotin tag linked via two different spacers to the adrenomedullin peptide sequence. A mix of panning rounds using non-specifically bound antigen and streptavidin bound antigen were used to minimize background of non-specific binders. The eluted phages from the third round of panning have been used for the generation of monoclonal scFv expressing E.coli strains. Supernatant from the cultivation of these clonal strains has been directly used for an antigen ELISA testing (Hust, M., Meyer, T., Voedisch, B., Riilker, T„ Thie, H„ El-Ghezal, A., Kirsch, M.I., Schiitte, M., Helmsing, S., Meier, D., Schirrmann, T., Diibel, S., 2011. A human scFv antibody generation pipeline for proteome research. Journal of Biotechnology 152, 159-170; Schiitte, M., Thullier, P., Pelat, T,, Wezler, X., Rosenstock, P., Hinz, D., Kirsch, M,I.,Hasenberg, M., Frank, R., Schirnnann, T., Gunzer, M., Hust, M., Diibel, S., 2009. Identification of a putative Crf splice variant and generation of recombinant antibodies for the specific detection of Aspergillus fumigatus. PLoS One 4, e6625).
Humanization of murine antibodies may be conducted according to the following procedure: For humanization of an antibody of murine origin the antibody sequence is analyzed for the structural interaction of framework regions (FR) with the complementary determining regions (CDR) and the antigen. Based on structural modelling an appropriate FR of human origin is selected and the murine CDR sequences are transplanted into the human FR. Variations in the amino acid sequence of the CDRs or FRs may be introduced to regain structural interactions, which were abolished by the species switch for the FR sequences. This recovery of structural interactions may be achieved by random approach using phage display libraries or via directed approach guided by molecular modeling (Almagro JC, Fransson J., 2008. Humanization of antibodies. Front Biosci. 2008 Jan 1 ;13:1619-33.).
In a preferred embodiment the ADM antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, F(ab)2 fragment and scFv-Fc Fusion protein. In another preferred embodiment the antibody format is selected from the group comprising scFab fragment, Fab fragment, scFv fragment and bioavailability optimized conjugates thereof, such as PEGylated fragments. One of the most preferred formats is scFab format. WO 2013/072513 PCT/EP2012/072932 19
In another preferred embodiment, the anti-ADM antibody, anti-ADM antibody fragment, or anti-ADM non-Ig scaffold is a full length antibody, antibody fragment, or non-Ig scaffold.
In a preferred embodiment the anti-adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is directed to and can bind to an epitope of at least 5 amino acids in length contained in ADM.
In another preferred embodiment the anti-adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is directed to and can bind to an epitope of at least 4 amino acids in length contained in ADM.
In one specific embodiment of the invention the anti-AdrenomeduIlin (ADM) antibody or anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin is provided for use in therapy of an acute disease or acute condition of a patient wherein said antibody or fragment or scaffold is not ADM-binding-Protein-1 (complement factor H).
In one specific embodiment of the invention the anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin is provided for use in therapy of an acute disease or acute condition of a patient wherein said antibody or antibody fragment or non-Ig scaffold binds to a region of preferably at least 4, or at least 5 amino acids within the sequence of aa 1-42 of mature human ADM: SEQ ID No.: 24 YRQSMNNFQGLRSFGCRFGTCTVQKLAHQIYQFTDKDKDNVA.
In one specific embodiment of the invention the anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin is provided for use in therapy of an acute disease or acute condition of a patient wherein said antibody or fragment or scaffold binds to a region of preferably at least 4, or at least 5 amino acids within the sequence of aa 1-21 of mature human ADM: WO 2013/072513 PCT/EP2012/072932 20 SEQ ID No.: 23 YRQSMNNFQGLRSFGCRFGTC.
In a preferred embodiment of the present invention said anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold binds to a region of ADM that is located in the N-terminal part (aa 1-21) of adrenomedullin, (see Fig. 2).
In another preferred embodiment said anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold recognizes and binds to the N-terminal end (aal) of adrenomedullin. N-terminal end means that the amino acid 1, that is “Y” of SEQ ID No. 21 or 23; is mandatory for antibody binding. Said antibody or fragment or non-Ig scaffold would neither bind N-terminal extended nor N-terminal modified Adrenomedullin nor N-terminal degraded adrenomedullin.
In a preferred embodiment the anti-adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold is directed to and can bind to an epitope of at least 5 amino acids in length contained in ADM, preferably in human ADM.
In a preferred embodiment the anti-adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold is directed to and can bind to an epitope of at least 4 amino acids in length contained in ADM, preferably in human ADM.
In another specific embodiment pursuant to the invention the herein provided anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold does not bind to die C-terminal portion of ADM, i.e. the aa 43 - 52 of ADM PRSKISPQGY-NH2 (SEQ ID NO: 25).
In one specific embodiment it is preferred to use an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention, wherein said anti-adrenomedullin antibody or said anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-Ig scaffold that enhances WO 2013/072513 PCT/EP2012/072932 21 the half life (ti/2; half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100%.
The half life (half retention time) of ADM may be determined in human plasma in absence and presence of an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-Ig scaffold, respectively, using an immunoassay for the quantification of ADM.
The following steps may be conducted: - ADM may be diluted in human citrate plasma in absence and presence of an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non~IG scaffold, respectively,and may be incubated at 24
°C
- Aliquots are taken at selected time points (e.g. within 24 hours) and degradation of ADM may be stopped in said aliquots by freezing at -20 °C - The quantity of ADM may be detennined by a hADM immunoassay directly, if the selected assay is not influenced by the stabilizing antibody. Alternatively, the aliquot may be treated with denaturing agents (like HCI) and, after clearing the sample (e.g. by centrifugation) the pH can be neutralized and the ADM-quantified by an ADM immunoassay. Alternatively, non-immunoassay technologies (e.g. rpHPLC) can be used for ADM-quantification. - The half life of ADM is calculated for ADM incubated in absence and presence of an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-lG scaffold, respectively, - The enhancement of half life is calculated for the stabilized ADM in comparison to ADM that has been incubated in absence of an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-IG scaffold. A two-fold increase of the half life of ADM is an enhancement of half life of 100%.
Half Life (half retention time) is defined as the period over which the concentration of a specified chemical or drug takes to fall to half baseline concentration in the specified fluid or blood. WO 2013/072513 PCT/EP2012/072932 22
An assay that may be used for the determination of the Half life (half retention time) of adrenomedullin in serum, blood, plasma is described in Example 3.
For other diseases blocking of ADM may be beneficial to a certain extent. However, it might also be detrimental if ADM is totally neutralized as a certain amount of ADM may be required for several physiological functions. In many reports it was emphasized that the administration of ADM may be beneficial in certain diseases. In contrast thereto in other reports ADM was reported as being life threatening when administered in certain conditions.
In a specific embodiment said anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold is a non-neutralizing antibody, fragment or non-Ig scaffold. A neutralizing anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold would block the bioactivity of ADM to nearly 100%, to at least more than 90%, preferably to at least more than 95%.
In contrast, a non-neutralizing anti-ADM antibody, or anti-ADM antibody fragment or anti-ADM non-Ig scaffold blocks the bioactivity of ADM less than 100%, preferably to less than 95%, preferably to less than 90%, more preferred to less than 80 % and even more preferred to less than 50 %. This means that the residual bioactivity of ADM bound to the nonneutralizing anti-ADM antibody, or anti-ADM antibody fragment or anti-ADM non-Ig scaffold would be more than 0%, preferably more than 5 %, preferably more than 10 % , more preferred more than 20 %, more preferred more than 50 %.
In this context (a) molecule(s), being it an antibody, or an antibody fragment or a non-Ig scaffold with “non-neutralizing anti-ADM activity”, collectively termed here for simplicity as “non-neutralizing” anti-ADM antibody, antibody fragment, or non-Ig scaffold, that e.g. blocks the bioactivity of ADM to less than 80 %, is defined as
a molecule or molecules binding to ADM, which upon addition to a culture of an eukaryotic cell line, which expresses functional human recombinant ADM receptor composed of CRLR (calcitonin receptor like receptor) and RAMP3 (receptor-activity modifying protein 3), reduces the amount of cAMP produced by the cell line through the action of parallel added human synthetic ADM WO 2013/072513 PCT/EP2012/072932 23 peptide, wherein said added human synthetic ADM is added in an amount that in the absence of the non-neutralizing antibody to be analyzed, leads to half-maximal stimulation of cAMP synthesis, wherein the reduction of cAMP by said molecule(s) binding to ADM takes place to an extent, which is not more than 80%, even when the non-neutralizing molecule(s) binding to ADM to be analyzed is added in an amount, which is 10-fold more than the amount, which is needed to obtain the maximal reduction of cAMP synthesis obtainable with the non-neutralizing antibody to be analyzed.
The same definition applies to the other ranges; 95%, 90%, 50% etc.
In a specific embodiment according to the present invention an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is used, wherein said antibody or antibody fragment or non-Ig scaffold blocks the bioactivity of ADM to less than 80 %, preferably less than 50% (of baseline values).
This is in the sense of blocking the circulating ADM of not more than 80% or not more than 50%, respectively.
It has been understood that said limited blocking of the bioactivity of ADM occurs even at excess concentration of the antibody, antibody fragment or non-Ig scaffold, meaning an excess of the antibody, antibody fragment or non-Ig scaffold in relation to ADM. Said limited blocking is an intrinsic property of the ADM binder itself. This means that said antibody, antibody fragment or non-Ig scaffold has a maximal inhibition of 80% or 50%, respectively. By implication, this means that 20% or 50% residual ADM bioactivity remains present although appropriate amounts or excess amounts of antibody, antibody fragment or non-Ig scaffold are administered, respectively.
In a preferred embodiment said anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold would block the bioactivity of ADM at least 5 %. By implication, this means residual 95% circulating ADM bioactivity remains present. This is the lower threshold of bioactivity remaining after administration of said anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold. The bioactivity is defined as the effect that a substance takes on a living organism or tissue or organ or functional unit in vivo or in vitro (e.g. in an assay) after its interaction. In case of ADM bioactivity this may be the effect of ADM in a human recombinant Adrenomedullin receptor cAMP functional assay. Thus, WO 2013/072513 PCT/EP2012/072932 24 according to the present invention bioactivity is defined via an Adrenomedullin receptor cAMP functional assay. The following steps may be performed in order to determine the bioactivity of ADM in such an assay: - Dose response curves are performed with ADM in said human recombinant Adrenomedullin receptor cAMP functional assay. - The ADM-concentration of half-maximal cAMP stimulation may be calculated. - At constant half-maximal cAMP-stimulating ADM-concentrations dose response curves (up to 100 pg/ml final concentration) are performed by an ADM stabilizing antibody or an adrenomedullin stabilizing antibody fragment or an adrenomedullin stabilizing non-Ig scaffold, respectively. A maximal (at maximal dose) inhibition by said ADM stabilizing antibody of 50% means that said ADM antibody or said adrenomedullin antibody fragment or said adrenomedullin non-Ig scaffold, respectively, blocks the bioactivity to 50% of baseline values. A maximal inhibition in said ADM bioassay of 80% means that said anti-ADM antibody or said anti-adrenomedullin antibody fragment or said anti-adrenomedullin non-Ig scaffold, respectively, blocks the bioactivity of ADM to 80%. This is in the sense of blocking the ADM bioactivity to not more than 80%.
The bioactivity of ADM may be determined in a human recombinant Adrenomedullin receptor cAMP functional assay (Adrenomedullin Bioassay) according to Example 2.
In a preferred embodiment a modulating anti-ADM antibody or a modulating anti-adrenomedullin antibody fragment or a modulating anti-adrenomedullin non-Ig scaffold is used in therapy of an acute disease or acute condition of a patient for stabilizing the circulation.
Such a modulating anti-ADM antibody or a modulating anti-ADM adrenomedullin antibody fragment or a modulating anti-ADM adrenomedullin non-Ig scaffold may be especially useful in the treatment of sepsis. A modulating anti-ADM antibody or a modulating anti-adrenomedullin antibody fragment or a modulating anti-adrenomedullin non-Ig scaffold enhances the bioactivity of ADM in the early phase of sepsis and reduces the damaging effects of ADM in the late phase of sepsis. WO 2013/072513 PCT/EP2012/072932 25 A “modulating” anti-ADM antibody or a modulating anti-adrenomedullin antibody fragment or a modulating anti-adrenomedullin non-Ig scaffold is an antibody or an adrenomedullin antibody fragment or non-Ig scaffold that enhances the half life (tm half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably >50 %, most preferably >100% and blocks the bioactivity of ADM to less than 80 %, preferably less than 50 % and wherein said anti-ADM antibody, anti-ADM antibody fragment or anti-ADM non-Ig scaffold would block the bioactivity of ADM at least 5 %. These values related to half-life and blocking of bioactivity have to be understood in relation to the before-mentioned assays in order to determine these values. This is in the sense of blocking the circulating ADM of not more than 80% or not more than 50%, respectively. This means 20% residual ADM bioactivity remains present, or 50% residual ADM bioactivity remains present, respectively.
Such a modulating anti-ADM antibody or a modulating anti-adrenomedullin antibody fragment or a modulating anti-adrenomedullin non-Ig scaffold offers the advantage that the dosing of the administration is facilitated. The combination of partially blocking or partially reducing Adrenomedullin bioactivity and increase of the in vivo half life (increasing the Adrenomedullin bioactivity) leads to beneficial simplicity of anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-adrenomedullin non-Ig scaffold dosing. In a situation of excess endogenous Adrenomedullin (maximal stimulation, late sepsis phase, shock, hypodynamic phase) the activity lowering effect is the major impact of the antibody or fragment or scaffold, limiting the (negative) effect of Adrenomedullin. In case of low or normal endogenous Adrenomedullin concentrations, the biological effect of anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is a combination of lowering (by partially blocking) and increase by increasing the Adrenomedullin half life, if the half life effect is stronger than the net blocking effect, the biological activity of endogenous Adrenomedullin is beneficially increased in early phases of sepsis (low Adrenomedullin, hyperdynamic phase). Thus, the non-neutralizing and modulating Adrenomedullin antibody or adrenomedullin antibody fragment or adrenomedullin non-Ig scaffold acts like an ADM bioactivity buffer in order to keep the bioactivity of ADM within a certain physiological range. WO 2013/072513 PCT/EP2012/072932 26
Thus, the dosing of the anti-ADM antibody/ffagment/scaffold in e.g. sepsis may be selected from an excessive concentration, because both sepsis phases (early and late) benefit from excessive anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold treatment in case of a modulating effect. Excessive means: The anti Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold concentration is higher than endogenous Adrenomedullin during late phase (shock) of e.g. sepsis. This means, in case of a modulating anti-ADM antibody or modulating anti-ADM antibody fragment or modulating anti-ADM non-Ig scaffold dosing in sepsis may be as follows:
The concentration of Adrenomedullin in septic shock is 226+/-66 fmol/ml (Nishio et al., "Increased plasma concentrations of adrenomedullin correlate with relaxation of vascular tone in patients with septic shock.", Crit Care Med. 1997, 25(6):953-7), an equimolar concentration of anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold is 42.5 pg/1 blood, (based on 6 1 blood volume / 80kg body weight) 3.2 pg/kg body weight. Excess means at least double (mean) septic shock Adrenomedullin concentration, at least > 3pg anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold / kg body weight, preferred at least 6.4pg anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold! kg body weight. Preferred > 10pg/kg, more preferred > 20pg/kg, most preferred > 100 pg anti-Adrenomedullin antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold / kg body weight.
This may apply to other severe and acute conditions than septic shock as well.
In a specific embodiment of the invention the anti-ADM antibody is a monoclonal antibody or a fragment thereof. In one embodiment of the invention the anti-ADM antibody or the anti-ADM antibody fragment is a human or humanized antibody or derived therefrom. In one specific embodiment one or more (murine) CDR’s are grafted into a human antibody or antibody fragment.
Subject matter of the present invention in one aspect is a human CDR-grafted anti-ADM antibody or anti-ADM antibody fragment thereof that binds to ADM, wherein the human CDR-grafted antibody or antibody fragment thereof comprises an antibody heavy chain (H chain) comprising WO 2013/072513 PCT/EP2012/072932 27
SEQ ID NO:l GYTFSRYW
SEQ ID NO: 2 ILPGSGST and/or
SEQ ID NO: 3 TEGYEYDGFDY and /or further comprises an antibody light chain (L chain) comprising:
SEQ ID NO:4 QSIVYSNGNTY
SEQ ID NO: 5 RVS and/or SEQ ID NO: 6 FQGSHIPYT.
In one specific embodiment of the invention subject matter of the present invention is a human monoclonal antibody that binds to ADM or an antibody fragment thereof wherein the heavy chain comprises at least one CDR selected from the group comprising:
SEQ ID NO: 1 GYTFSRYW
SEQ ID NO: 2 ILPGSGST WO 2013/072513 PCT/EP2012/072932 28
SEQ ID NO: 3 TEGYEYDGFDY and wherein the light chain comprises at least one CDR selected from the group comprising:
SEQ ID No: 4 QSIVYSNGNTY
SEQ ID NO: 5 RVS SEQ ID NO: 6 FQGSHIPYT.
In a more specific embodiment of the invention subject matter of the invention is a human monoclonal antibody that binds to ADM or an antibody fragment thereof wherein the heavy chain comprises the sequences
SEQ ID NO: 1 GYTFSRYW
SEQ ID NO: 2 ILPGSGST
SEQ ID NO: 3 TEGYEYDGFDY and wherein the light chain comprises the sequences
SEQ ID NO: 4 QSIVYSNGNTY
SEQ ID NO: 5 RVS WO 2013/072513 PCT/EP2012/072932 29 SEQ ID NO: 6 FQGSHIPYT.
In a very specific embodiment the anti-ADM antibody has a sequence selected from the group comprising: SEQ ID NO 7, 8, 9,10,11, 12, 13 and 14.
The anti-ADM antibody or anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention exhibits an affinity towards human ADM in such that affinity constant is greater than 10’7 M, preferred 10~8 M, preferred affinity is greater than 10‘9 M, most preferred higher than 10‘10 M. A person skilled in the art knows that it may be considered to compensate lower affinity by applying a higher dose of compounds and this measure would not lead out-of-the-scope of the invention. The affinity constants may be determined according to the method as described in Example 1.
In a preferred embodiment the antibody or the antibody fragment is used for reducing the risk of mortality during said chronic or acute disease of a patient wherein said disease is selected from the group comprising sepsis, diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction.
In a preferred embodiment the anti-ADM antibody or the anti-ADM antibody fragment or anti-ADM non-Ig scaffold is used for reducing the risk of mortality during said acute disease or acute condition of a patient.
It should be emphasized that the patient having an acute disease or acute condition may be characterized by need for stabilizing the circulation meaning the systemic circulation or by the need for preventively stabilizing the circulation meaning the systemic circulation.
Chronic or acute disease or acute condition according to the present invention may be a disease or condition selected from the group comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrome (SIRS) sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, WO 2013/072513 PCT/EP2012/072932 30 liver dysfunction, burnings, surgery, traumata, poisoning, damages by chemotherapy. Especially useful is the antibody or fragment or scaffold according to the present invention for reducing the risk of mortality during sepsis and septic shock, i.e. late phases of sepsis.
However, it should be emphasized that the medicaments provided by the present invention, being anti-ADM antibodies, anti-ADM antibody fragments, or anti-ADM non-Ig scaffolds are only intended to be used for sake of stabilizing the systemic circulation in a patient in need for stabilizing the systemic circulation or by the need for preventively stabilizing the systemic circulation, and thus not for any methods of primary treatment to a chronic or acute disease or condition itself. This means the present invention does not provide for a therapy of healing/curing e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS), or sepsis, or severe sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata, poisoning, or damages induced by chemotherapy within the scope of the invention.
In one embodiment the anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is used in therapy of acute disease or acute condition of a patient according to the present invention, wherein said patient is an ICU patient. In another embodiment the anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold is used in therapy of acute disease of a patient according to the present invention, wherein said patient is critically ill. Critically ill means the patient is having a disease or state in which death is possible or imminent.
Subject of the present invention is further an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold for use in therapy of acute disease of a patient according to the present invention, wherein said antibody or antibody fragment or non-Ig scaffold is to be used in combination of ADM binding protein. ADM binding protein is also naturally present in the circulation of said patient.
It should be emphasized that the term “ADM binding protein” comprises ADM-binding-protein-1 (complement factor H). However, said ADM binding protein by definition pursuant to the invention is neither a non-neutralizing anti-ADM antibody/antibody fragment/non-Ig scaffold nor a modulating anti-ADM antibody/antibody fragment/non-Ig scaffold. PCT/EP2012/072932 WO 2013/072513 31
Subject of the present invention is further an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-fg scaffold for use in therapy of acute disease or acute condition of a patient according to the present invention, wherein said antibody or antibody fragment or non-Ig scaffold may be used in combination with further active ingredients.
Subject matter of the invention is also an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold may be used in combination with a primary medicament, wherein said combination is for use in therapy of a acute disease or acute condition of a patient for stabilizing the circulation of said patient.
In this regard, it should be emphasized that the anti-ADM antibody/antibody ffagment/non-Ig scaffold are not to be administered as primary medicament or as first-line-treatment of any underlying disease or condition, irrespective of being acute or chronic, but administration of said anti-ADM antibody/antibody fragment/non-Ig scaffold pursuant to the invention is to be intended for patients with acute disease or acute condition associated with weak circulation or circulation problems, and thus who are in need for stabilizing the circulation.
Primary medicament means a medicament that acts against the primary cause of said disease or condition. Said primary medicament may be antibiotics in case of infections.
In a specific embodiment of the before mentioned combinations said combinations are to be used in combination with vasopressors e.g. catecholamine wherein said further combination is for use in therapy of a chronic or acute disease or condition of a patient for stabilizing the circulation.
In one embodiment of the invention said patient having a chronic or acute disease or chronic condition being in need for stabilizing the circulation is characterized by the need of the patient to get administration of vasopressors e.g. catecholamine administration.
Subject matter of the invention in one specific embodiment is, thus, an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or an anti-ADM non-Ig scaffold to be used in combination with ADM binding protein and/or further active ingredients for use in therapy of a patient in need of a treatment of vasopressors e.g. catecholamine. WO 2013/072513 PCT/EP2012/072932 32
In a specific embodiment of the above mentioned combinations said combinations are to be used in combination with fluids administered intravenously, wherein said combination is for use in therapy of a chronic or acute disease or condition of a patient for stabilizing the circulation. 5 In one embodiment of the invention said patient having a chronic or acute disease or acute condition being in need for stabilizing the circulation is characterized by the need of the patient to get intravenous fluids.
Subject matter of the invention in one specific embodiment is, thus, an anti-Adrenomedullin (ADM) antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold 10 in combination with ADM binding protein and/or further active ingredients for use in therapy of a patient in need of intravenous fluids.
Said anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold or combinations thereof with ADM binding protein and/or further active ingredients may be used in combination with catecholamine and/or with fluids administered 15 intravenously for use in a method of treating acute disease or acute condition of a patient for stabilizing the circulation.
Subject matter of the invention is also an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention to be used 20 in combination with TNF-alpha-antibodies. TNF-alpha-antibodies are commercially available for the treatment of patients.
Subject matter of the invention is also an anti-ADM antibody or an anti-adrenomedullin antibody fragment or anti-ADM non-Ig scaffold according to the present invention to be used 25 in combination with antibiotics.
Subject of the present invention is further a pharmaceutical formulation comprising an anti-ADM antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold according to the present invention. Subject of the present invention is further a pharmaceutical formulation 30 according to the present invention wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. In another embodiment subject of the present invention is further a pharmaceutical formulation according to the present invention wherein said pharmaceutical formulation is in a dried state to be reconstituted before use. WO 2013/072513 PCT/EP2012/072932 33
Said pharmaceutical formulation may be administered intra-muscular. Said pharmaceutical formulation may be administered intra-vascular. Said pharmaceutical formulation may be administered via infusion. In another embodiment subject of the present invention is further a pharmaceutical formulation according to the present invention wherein said pharmaceutical formulation is in a freeze-dried state.
It should be emphasized that the pharmaceutical formulation in accordance with the invention as may be administered intra-muscular, intra-vascular, or via infusion is preferably administered to a patient for stabilizing the systemic circulation with the proviso that said patient is in need for stabilizing the circulation.
Therefore, in another embodiment of the present invention the pharmaceutical formulation according to the present invention is to be administered to a patient for stabilizing the systemic circulation with the proviso that said patient is in need for stabilizing the circulation.
In another more preferred embodiment the present invention provides for a pharmaceutical formulation comprising an anti-Adrenomedullin (ADM) antibody or an anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig scaffold binding to adrenomedullin for use in therapy of an acute disease or acute condition of a patient, wherein said pharmaceutical formulation is to be administered to a patient for stabilizing the systemic circulation with the proviso that said patient is in need for stabilizing the systemic circulation.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronic or acute disease of a patient for the regulation of liquid balance. 2. ADM antibody or an adrenomedullin antibody fragment according to claim 1 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. WO 2013/072513 PCT/EP2012/072932 34 3. ADM antibody or an adrenomedullin antibody fragment according claim 1 or 2 wherein said antibody or fragment binds to the N-terminal part (aa 1-21) of adrenomedullin. 4. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 3, wherein said antibody or fragment recognizes and binds to the N-terminal end (aal) of adrenomedullin. 5. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 4, wherein said antibody or fragment is an ADM stabilizing antibody or ADM stabilizing a antibody fragment that enhances the ti/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100 %. 6. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 5, wherein said antibody or fragment blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 7. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronic or acute disease of a patient according to any of claims 1 to 6 wherein said disease is selected from the group comprising sepsis, diabetis, cancer, heart failure, shock and kidney dysfunction. 8. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronic or acute disease of a patient according to any of claims 1 to 7 wherein said patient is an ICU patient. 9. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronic or acute disease of a patient according to any of claims 1 to 7 wherein said antibody or fragment is a modulating antibody or fragment that enhances the tm half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100 % and that blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. WO 2013/072513 PCT/EP2012/072932 35 10. Pharmaceutical formulation comprising an antibody or fragment according to any of claims 1 to 9. 11. Pharmaceutical formulation according to claim 10 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 12. Pharmaceutical formulation according to claim 10 wherein said pharmaceutical formulation is in a freeze-dried state. 13. Pharmaceutical formulation according to any of claims 10 to 11, wherein said pharmaceutical formulation is administered intra-muscular. 14. Pharmaceutical formulation according to any of claims 10 to 11, wherein said pharmaceutical formulation is administered intra-vascular. 15. Pharmaceutical formulation according to claim 14, wherein said pharmaceutical formulation is administered via infusion.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin ADM antibody or an adrenomedullin antibody fragment an ADM non-Ig scaffold for use in therapy of a chronic or acute disease or acute condition of a patient for the regulation of fluid balance. 2. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to claim 1 wherein said ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold is a non-neutralizing ADM antibody or a nonneutralizing adrenomedullin antibody fragment or a non-neutralizing ADM non-IG scaffold. 3. Adrenomedullin ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold for use in therapy of a chronic or acute disease or acute condition according to claim 1 or 2 for preventing or reducing edema in said patient. WO 2013/072513 PCT/EP2012/072932 36 4. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 3 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. 5. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 4, wherein said antibody or fragment or scaffold binds to the N-terminal part (aa 1-21) of adrenomedullin. 6. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 5, wherein said antibody or fragment scaffold recognizes and binds to the N-terminal end (aal) of adrenomedullin, 7. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 6, wherein said antibody or fragment or scaffold is an ADM stabilizing antibody or ADM stabilizing antibody fragment or ADM stabilizing non-IG scaffold that enhances the half life (lm half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100 %. 8. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 7, wherein said antibody or fragment blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 9. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of claims 1 to 8 wherein said disease is selected from the group comprising SIRS, sepsis, diabetis, cancer, heart failure, shock and kidney dysfunction 10. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 9, wherein said antibody or fragment is a human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof wherein the heavy chain comprises the sequences SEQ ID NO: 1 WO 2013/072513 PCT/EP2012/072932 37
GYTFSRYW
SEQ ID NO: 2 ILPGSGST
SEQ ID NO: 3 TEGYEYDGFDY and wherein the light chain comprises the sequences 10 SEQ ID NO: 4
QSIVYSNGNTY
SEQ ID NO: 5 RVS SEQ ID NO: 6 FQGSHIPYT. 11. A human monoclonal antibody or fragment that binds to ADM or an antibody fragment 20 thereof according to claim 10 wherein said antibody or fragment comprises a sequence selected from the group comprising : SEQ ID NO: 7 (AM-VH-C)
QVQLQQSGAELMKPGASVKISCKATGYTFSRYWIEWVKQRPGHGLEWIGEIL 25 PGSGSTNYNEKFKGKATITADTSSNTAYMQLSSLTSEDSAVYYCTEGYEYDGF
D Y W GQGTTLTV S S ASTKGPS VFPLAPSSKSTSGGT AALGCLVKD YFPEP VTVS WN S GALT S G VHTFP A VLQS SGLY S LS SWT VPS S SLGT QTYICN VNFIKP SNTK VDKRVEPKHHHHHH WO 2013/072513 PCT/EP2012/072932 38 SEQ ID NO: 8 (AM-VH1)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWISWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WNSG ALTSG VHTFP A VLQS SGLY SLS SWT VP S S SLGTQT YICNVNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 9 (AM-VH2-E40)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWIEWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WN SG ALTS G VHTFP AVLQS SGLYSLS SWT VPS S SLGTQTYICN VNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWISWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WN S GALT S G VHTFP A VLQS S GL Y S LS SWT VP S S SLGTQT YICNVNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWIEWVRQAPGQGLEWMGEI
LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG
FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV
SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT
KVDKRVEPKHHHHHH SEQ ID NO: 12 (AM-VL-C)
DVLLSQTPLSLPVSLGDQATISCRSSQSIVYSNGNTYLEWYLQKPGQSPKLLIY RVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHIPYTFGGGTK LEIKRT V A AP SVFIFPPSDEQ LKS GT AS WCLLNNF YPRE AKV Q WKVDN ALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF NRGEC WO 2013/072513 PCT/EP2012/072932 39 SEQ ID NO: 13 (AM-VL1)
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLNWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQ S GNSQES VTEQDSKD STY S LS STLTLSKAD YEKHKV Y ACEVTHQGLS SPVTKS FNRGEC SEQ ID NO: 14 (AM-VL2-E40)
DVYMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLEWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRT VA AP SVFIFPPSDEQLKSGT AS V V C LLNNF YPRE AKV Q WKVDNALQ S GNSQES VTEQDSKD STY SLSSTLTLSKAD YEKHKVY ACEVTHQGLS SPVTKS
FNRGEC 12. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of claims 1 to 9 wherein said patient is an ICU patient. 13. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of claims 1 to 12 wherein said antibody or fragment or scaffold is a modulating antibody or fragment or scaffold that enhances the half life (ti/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100 % and that blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 14. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to 13 to be used in combination with catecholamine and/ or fluids administered intravenously. 15. ADM antibody or adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to WO 2013/072513 PCT/EP2012/072932 40 13 or a combination according to claim 12 to be used in combination with ADM binding protein and/or further active ingredients. 16. Pharmaceutical formulation comprising an antibody or fragment or scaffold according to any of claims 1 to 15. 17. Pharmaceutical formulation according to claim 16 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 18. Pharmaceutical formulation according to claim 16 wherein said pharmaceutical formulation is in a freeze-dried state. 19. Pharmaceutical formulation according to any of claims 16 to 17, wherein said pharmaceutical formulation is administered intra-muscular. 20. Pharmaceutical formulation according to any of claims 16 to 17, wherein said pharmaceutical formulation is administered intra-vascular. 21. Pharmaceutical formulation according to claim 20, wherein said pharmaceutical formulation is administered via infusion.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment for use in therapy of a chronic or acute disease of a patient for stabilizing the circulation. 2. ADM antibody or an adrenomedullin antibody fragment according to claim 1 wherein said antibody or fragment reduces the catecholamine requirement of said patient. 3. ADM antibody or an adrenomedullin antibody fragment according to claim 1 or 2 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. WO 2013/072513 PCT/EP2012/072932 41 4. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 3 wherein said antibody or fragment binds to the N-terminal part (aa 1-21) of adrenomedullin. 5. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 4, wherein said antibody or fragment recognizes and binds to the N-terminal end (aal) of adrenomedullin. 6. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 5, wherein said antibody or fragment is an ADM stabilizing antibody that enhances the tl/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably > 50 %, most preferably >100 %. 7. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 6, wherein said antibody or fragment blocks the bioactivity of ADM to less than 80 %, preferably less than 50 %. 8. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 7, wherein said antibody or fragment is a modulating ADM antibody or a modulating adrenomedullin antibody fragment that enhances the tl/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably > 50 %, most preferably >100 % and that blocks the bioactivity of ADM to less than 80 %, preferably less than 50 %: 9. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to 8 wherein said disease is selected from the group comprising sepsis, diabetis, cancer, acute and chronic vascular diseases as e.g. heart failure, shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction. 10. Pharmaceutical formulation comprising an antibody according to any of claims 1 to 9. 11. Pharmaceutical formulation according to claim 10 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. WO 2013/072513 PCT/EP2012/072932 42 12. Pharmaceutical formulation according to claim 10 wherein said pharmaceutical formulation is in a freeze-dried state. 13. Pharmaceutical formulation according to any of claims 10 to 11, wherein said pharmaceutical formulation is administered intra-muscular. 14. Pharmaceutical formulation according to any of claims 10 to 11, wherein said pharmaceutical formulation is administered intra-vascular. 15. Pharmaceutical formulation according to claim 14, wherein said pharmaceutical formulation is administered via infusion.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment or an ADM non-IG scaffold for use in therapy of a chronic or acute disease or condition of a patient for stabilizing the circulation. 2. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to claim 1 wherein said antibody or fragment or scaffold reduces the vasopressor requirement, e.g. catecholamine requirement of said patient, 3. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to claim 1 or 2 wherein said ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold is a non-neutralizing ADM antibody or a nonneutralizing adrenomedullin antibody fragment or a non-neutralizing ADM non-IG scaffold. 4. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 3 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. WO 2013/072513 PCT/EP2012/072932 43 5. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 4 wherein said antibody or fragment or scaffold binds to the N-terminal part (aa 1-21) of adrenomedullin. 6. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 5, wherein said antibody or fragment or scaffold recognizes and binds to the N-terminal end (aal) of adrenomedullin. 7. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 6, wherein said antibody or fragment or scaffold is an ADM stabilizing antibody or fragment or scaffold that enhances the half life (tl/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably > 50 %, most preferably >100 %. 8. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 7, wherein said antibody or fragment or scaffold blocks the bioactivity of ADM to less than 80 %, preferably less than 50 %. 9. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 8, wherein said antibody or fragment or scaffold is a modulating ADM antibody or a modulating adrenomedullin antibody fragment or scaffold that enhances the half life (tl/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably > 50 %, most preferably >100 % and that blocks the bioactivity of ADM to less than 80 %, preferably less than 50 %: 10. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 9, wherein said antibody or fragment is a human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof wherein the heavy chain comprises the sequences SEQ ID NO: 1
GYTFSRYW WO 2013/072513 PCT/EP2012/072932 44
SEQ ID NO: 2 ILPGSGST SEQ ID NO: 3
5 TEGYEYDGFDY and wherein the light chain comprises the sequences SEQ ID NO:4
QSIVYSNGNTY 10
SEQ ID NO: 5 RVS SEQ ID NO: 6 15 FQGSHIPYT. 11. A human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof according to claim 10 wherein said antibody or fragment comprises a sequence selected from the group comprising : 20 SEQ ID NO: 7 (AM-VH-C)
QVQLQQSGAELMKPGASVKISCKATGYTFSRYWIEWVKQRPGHGLEWIGEIL PGSGSTNYNEKFKGKATITADTSSNTAYMQLSSLTSEDSAVYYCTEGYEYDGF DYWGQGTTLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVS 25 WNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTK
VDKRVEPKHHHHHH SEQ ID NO: 8 (AM-VH1) WO 2013/072513 PCT/EP2012/072932 45
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWISWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WNS G ALTSG VHTFP A VLQ SSGLYSLSS V VT VP S S SLGTQT YICNVNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 9 (AM-VH2-E40)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWffiWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FD YW GQGTTVT VS S ASTKGPS VFPLAP S SKSTS GGTA ALGCLVKD YFPEP VTV SWNSG ALT SG VHTFP A VLQSSGLY SLS SWT VP S S SLGTQT YICNVNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWISWVRQAPGQGLEWMGEI LP G S GSTN Y AQKFQ GRVTIT ADEST ST AYMELS SLRSEDT AVY Y CTEG YEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWIEWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WN S G ALTS G VHTFP AVLQS S G LY SLS SWT VP S S S LGTQT YICNVNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 12 (AM-VL-C)
DVLLSQTPLSLPVSLGDQATISCRSSQSIVYSNGNTYLEWYLQKPGQSPKLLIY
RVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHIPYTFGGGTK
LEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQS
GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF
NRGEC SEQ ID NO: 13 (AM-VL1) WO 2013/072513 PCT/EP2012/072932 46
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLNWFQQRPGQSPRRLIY
RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT
KLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQ
SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS
FNRGEC SEQ ID NO: 14 (AM-VL2-E40)
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLEWFQQRPGQSPRRLIY
RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT
KLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQ
SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS
FNRGEC 12. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to 11 wherein said disease is selected from the group comprising SIRS, sepsis, diabetis, cancer, acute and chronic vascular diseases as e.g. heart failure, shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction. 13. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to 12 to be used in combination with catecholamine and/ or fluids administered intravenously. 14. ADM antibody or adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to 13 or a combination according to claim 10 to be used in combination with ADM binding protein and/or further active ingredients. 15. Pharmaceutical formulation comprising an antibody or fragment or non-IG scaffold according to any of claims 1 to 14. WO 2013/072513 PCT/EP2012/072932 47 16. Pharmaceutical formulation according to claim 15 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 17. Pharmaceutical formulation according to claim 15 wherein said pharmaceutical formulation is in a freeze-dried state. 18. Pharmaceutical formulation according to any of claims 15 to 16, wherein said pharmaceutical formulation is administered intra-muscular. 19. Pharmaceutical formulation according to any of claims 14 to 16, wherein said pharmaceutical formulation is administered intra-vascular. 20. Pharmaceutical formulation according to claim 16, wherein said pharmaceutical formulation is administered via infusion.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease wherein said antibody or said fragment is an ADM stabilizing antibody or fragment that enhances the ti/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably >50 %, most preferably 100 % and/or wherein said antibody blocks the bioactivity of ADM to less than 80 %, preferably to less than 50 %. 2. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease wherein said antibody or said fragment is a modulating ADM antibody or fragment that enhances the t)/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably >50 %, most preferably 100 % and that blocks the bioactivity of ADM to less than 80 %, preferably to less than 50 %. 3. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease according to claim 1 or 2, wherein said antibody or fragment binds to the N-terminal part (aa 1-21) of adrenomedullin. WO 2013/072513 PCT/EP2012/072932 48 4. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease wherein said antibody or said fragment according to claim 3 binds to the N-terminal end of adrenomedullin. 5. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in use in a treatment of a chronic or acute disease according to any of claims 1 to 4, wherein said antibody or said fragment is an ADM stabilizing antibody or fragment that enhances the t]/2 half retention time of adrenomedullm in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably >50 %, most preferably 100 %. 6. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease according to any of claims 1 to 5, wherein said antibody or said fragment blocks the bioactivity of ADM to less than 80 %, preferably to less than 50 %. 7. Adrenomedullin antibody or an adrenomedullin antibody fragment according to any of the claims 1 to 6 for use in a treatment of a chronic or acute disease wherein said disease is selected from the group comprising SIRS, sepsis, septic shock, diabetis, cancer, heart failure, shock, organ failure, kidney dysfunction, acute liquid dysbalance, and low blood pressure. 8. Adrenomedullin antibody or an adrenomedullin antibody fragment according to any of the claims 1 to 7 for use in a treatment of a chronic or acute disease wherein said disease is septic shock or sepsis. 9. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease according to any of the claims 1 to 8 wherein said antibody or fragment regulates the liquid balance of said patient. 10. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease according to any of the claims 1 to 9 wherein said antibody or fragment used for prevention of organ dysfunction or organ failure. 11. Adrenomedullin antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease according to claim 10 wherein said antibody or fragment is used for prevention of kidney dysfunction or kidney failure. WO 2013/072513 PCT/EP2012/072932 49 12. Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease in a patient according to claims 1 to 11 wherein said antibody or fragment is used for stabilizing the circulation. 13. ADM antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease in a patient according to claim 12 wherein said antibody or fragment reduces the catecholamine requirement of said patient. 14. ADM antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease in a patient according to any of claims 1 to 13 for the reduction of the mortality risk for said patient. 15. ADM antibody or an adrenomedullin antibody fragment for use in a treatment of a chronic or acute disease in a patient according to any of claims 1 to 14 wherein said antibody or fragment may be administered in a dose of at least 3 pg / Kg body weight. 16. Pharmaceutical composition comprising an antibody or fragment according to any of claims 1 to 15.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold wherein said antibody or said fragment or scaffold is a non-neutralizing antibody. 2. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold wherein said antibody or said fragment or scaffold is an ADM stabilizing antibody or fragment or scaffold that enhances the half life (ti/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably 100 % and/or wherein said antibody or fragment or scaffold blocks the bioactivity of ADM to less than 80 %, preferably to less than 50 %. 3. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-lg scaffold wherein said antibody or said fragment is a modulating ADM antibody or fragment or scaffold that enhances the half life (ti/2 half retention time) of adrenomedullin in scrum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably >50 %, most preferably 100 % and that blocks the bioactivity of ADM to less than 80 %, preferably to less than 50 %. WO 2013/072513 PCT/EP2012/072932 50 4. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to claim 1 or 2, wherein said antibody or fragment or scaffold binds to the N-terminal part (aa 1-21) of adrenomedullin. 5. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold wherein said antibody or said fragment or scaffold according to claim 3 binds to the N-terminal end of adrenomedullin. 6. Adrenomedullin antibody or an adrenomedullin antibody fragment ADM non-Ig scaffold according to any of claims 1 to 4, wherein said antibody or said fragment or said scaffold is an ADM stabilizing antibody or fragment that enhances the tm half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably >50 %, most preferably 100 %. 7. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 6 for use as an active pharmaceutical substance, 8. Adrenomedullin antibody or an adrenomedullin antibody fragment ADM non-Ig scaffold according to any of the claims 1 to 7 for use in a treatment of a chronic or acute disease or acute condition wherein said disease or condition is selected from the group comprising severe infections as e.g. meningitis, systemic inflammatory Response-Syndrome (SIRS,) sepsis; other diseases as diabetes, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction, burnings, surgery, traumata. 9. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 8 for use in a treatment of a chronic or acute disease or acute condition wherein said disease is septic shock or sepsis. 10. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 9, wherein said antibody or fragment is a human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof wherein the heavy chain comprises at least one of the sequences : SEQ ID NO: 1
GYTFSRYW WO 2013/072513 PCT/EP2012/072932 51
SEQ ID NO: 2 ILPGSGST
SEQ ID NO: 3 TEGYEYDGFDY
And/or wherein the light chain comprises the at least one of the sequences SEQ ID NO:4
QSIVYSNGNTY
SEQ ID NO: 5 RVS SEQ ID NO: 6 FQGSHIPYT. 11. A human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof according to claim 10 wherein said antibody or fragment comprises a sequence selected from the group comprising: SEQ ID NO: 7 (AM-VH-C)
QVQLQQSGAELMKPGASVKISCKATGYTFSRYWIEWVKQRPGHGLEWIGEIL
PGSGSTNYNEKFKGKATITADTSSNTAYMQLSSLTSEDSAVYYCTEGYEYDGF
DYWGQGTTLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTK
VDKRVEPKHHHHHH SEQ ID NO: 8 (AM-VH1) WO 2013/072513 PCT/EP2012/072932 52
QV QLVQSGAEVKKPGS S VKV SCKAS GYTFSRY WIS WVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTYTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WNSGALTS GVHTFP A VLQS SGLY S LS S VVTVPS S SLGTQTYICNVNHKPSNT KVDKRVEP KHHHHHH SEQ ID NO: 9 (AM-VH2-E40)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWIEWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FD YW GQGTTVTV S S ASTKGP S VFPLAP SSKSTSGGTAALGCLVKD YFPEPVTV S WN SG ALTSG VHTFP A VLQS SGLY S LS SWT VP S S SLGTQTYICNVNFIKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWISWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV S WNS G ALTS GVHTFP AVLQS SGLY S LS SWT VPS S SLGTQTYICNVNFIKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWIEWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSS ASTKGP S VFPLAP SSKSTSGGT A ALGCLVKD YFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 12 (AM-VL-C)
DVLLSQTPLSLPVSLGDQATISCRSSQSIVYSNGNTYLEWYLQKPGQSPKLLIY
RVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHIPYTFGGGTK
LEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS
GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF
NRGEC SEQ ID NO: 13 (AM-VL1) WO 2013/072513 PCT/EP2012/072932 53
DWMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLNWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQ S GN SQESVTEQD SKD S T YS LS STLT LSKAD YEKHKV Y AC E VTHQGLSSP VTKS
FNRGEC SEQ ID NO: 14 (AM-VL2-E40)
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLEWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQ SGNSQESVTEQDSKDSTY SLSSTLTLSKADYEKHKVY ACEYTHQGLSSPVTKS
FNRGEC 12. Adrenoraedullin antibody or an adrenomednllin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 11 for regulating the fluid balance in a patient having a chronic or acute disease or acute condition. . 13. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 11 for preventing or reducing organ dysfunction or organ failure in a patient having in a chronic or acute disease or acute condition. 14. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to claim 10 wherein organ is kidney or liver. 15. Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to claims 1 to 14 for stabilizing the circulation in a patient having a chronic or acute disease or acute condition. 16. ADM antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold for use in a treatment of a chronic or acute disease in a patient according to claim 15 wherein said antibody or fragment reduces the catecholamine requirement of said patient. 17. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 16 to be used in combination with vasopressors e.g. catecholamine. WO 2013/072513 PCT/EP2012/072932 54 18. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 17 to be used in combination with intravenous fluid administration. 19. Adrenomedullin antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold according to any of the claims 1 to 18 to be used in combination with an TNF-alpha-antibody. 20. ADM antibody or an adrenomedullin antibody fragment or non-Ig-scaffold according to any of claims 1 to 19 for use in a treatment of a patient in need thereof wherein said antibody or fragment may be administered in a dose of at least 3 pg / Kg body weight. 21. Pharmaceutical composition comprising an antibody or fragment or scaffold according to any of claims 1 to 20. 22. ADM antibody or an adrenomedullin antibody fragment or non-Ig-scaffold according to any of claims 1 to 20 for use in a treatment of a chronic or acute disease or chronic condition. 23. ADM antibody or an adrenomedullin antibody fragment or non-Ig-scaffold according to claim 22 wherein said disease is sepsis.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin ADM antibody or an adrenomedullin antibody fragment for use in therapy of a severe chronical or acute disease of a patient for the reduction of the mortality risk for said patient. 2. ADM antibody or an adrenomedullin antibody fragment according to claim 1 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. 3. ADM antibody or an adrenomedullin antibody fragment according claim 1 or 2 wherein said antibody or fragment binds to the N-terminal part (aa 1-21) of adrenomedullin. WO 2013/072513 PCT/EP2012/072932 55 4. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 3, wherein said antibody or fragment recognizes and binds to the N-terminal end (aal) of adrenomedullin. 5. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 4, wherein said antibody or fragment is an ADM stabilizing antibody or fragment that enhances the tl/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably > 50 %, most preferably >100 %. 6. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 5, wherein said antibody or fragment blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 7. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 6 wherein said disease is selected from the group comprising sepsis, diabetis, cancer, heart failure, shock and kidney dysfunction. 8. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 7 wherein said patient is an ICU patient. 9. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 8 wherein the mortality risk is reduced by preventing adverse event wherein the latter are selected from the group comprising SIRS, sepsis, septic shock, organ failure, kidney failure, liquid dysbalance and low blood pressure. 10. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 8 wherein said antibody or fragment is to be used in combination of ADM binding protein. WO 2013/072513 PCT/EP2012/072932 56 11. Pharmaceutical formulation comprising an antibody or fragment according to any of claims 1 to 10. 12. Pharmaceutical formulation according to claim 11 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 13. Pharmaceutical formulation according to claim 11 wherein said pharmaceutical formulation is in a freeze-dried state. 14. Pharmaceutical formulation according to any of claims 11 to 12, wherein said pharmaceutical formulation is administered intra-muscular. 15. Pharmaceutical formulation according to any of claims 11 to 12, wherein said pharmaceutical formulation is administered intra-vascular. 16. Pharmaceutical formulation according to claim 15, wherein said pharmaceutical formulation is administered via infusion.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold for use in therapy of a severe chronical or acute disease or acute condition of a patient for the reduction of the mortality risk for said patient wherein said antibody or fragment or scaffold is a non-neutralizing ADM antibody or a nonneutralizing adrenomedullin antibody fragment or a non-neutralizing ADM non-Ig scaffold. 2. ADM antibody or an adrenomedullin antibody fragment according to claim 1 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. 3. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold according claim 1 or 2 wherein said antibody or fragment or scaffold binds to the N-terminal part (aa 1-21) of adrenomedullin. WO 2013/072513 PCT/EP2012/072932 57 4. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold according to any of claims 1 to 3, wherein said antibody or fragment or scaffold recognizes and binds to the N-terminal end (aal) of adrenomedullin. 5. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold according to any of claims 1 to 4, wherein said antibody or fragment or scaffold is an ADM stabilizing antibody or fragment or scaffold that enhances the half life (tl/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least, 50 %, more preferably > 50 %, most preferably >100%. 6. ADM antibody or an adrenomedullin antibody fragment or an ADM non-lg scaffold according to any of claims 1 to 5, wherein said antibody or fragment or scaffold blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 7. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 6 wherein said disease is selected from the group comprising severe infections as e.g. meningitis, Systemic inflammatory Response-Syndrom (SIRS,) sepsis; other diseases as diabetis, cancer, acute and chronic vascular diseases as e.g. heart failure, myocardial infarction, stroke, atherosclerosis; shock as e.g. septic shock and organ dysfunction as e.g. kidney dysfunction, liver dysfunction; burnings, surgery, traumata. 8. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 7 wherein said disease is selected from the group comprising SIRS, a severe infection, sepsis, shock e.g.septic shock . 9. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold for use in therapy of a chronical or acute disease or acute condition of a patient according to any of claims 1 to 8 wherein said patient is an ICU patient. ADM antibody or an adrenomedullin antibody fragment or an ADM non-Ig scaffold for use in therapy of a chronical or acute disease or acute condition of a patient according to any of claims 1 to 9 wherein the mortality risk is reduced by preventing an adverse WO 2013/072513 PCT/EP2012/072932 58 event wherein the latter are selected from the group comprising SIRS, sepsis, shock as e.g, septic shock, acute and chronic vascular diseases as e.g. acute heart failure, myocardial infarction, stroke; organ failure as e.g, kidney failure, liver failure, fluid dysbalance and low blood pressure. 10. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 9, wherein said antibody or fragment is a human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof wherein the heavy chain comprises the sequences SEQ ID NO: 1
GYTFSRYW SEQ ID NO: 2 ILPGSGST SEQ ID NO: 3 TEGYEYDGFDY and wherein the light chain comprises the sequences SEQ ID NO:4
QSIVYSNGNTY
SEQ ID NO: 5 RVS SEQ ID NO: 6 FQGSHIPYT. WO 2013/072513 PCT/EP2012/072932 59 12. A human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof according to claim 10 wherein said antibody or fragment comprises a sequence selected from the group comprising : 5 SEQ ID NO: 7 (AM-VH-C)
QVQLQQSGAELMKPGASVKISCKATGYTFSRYWIEWVKQRPGHGLEWIGEIL PGSGSTNYNEKFKGKATITADTSSNTAYMQLSSLTSEDSAVYYCTEGYEYDGF D Y W GQGTTLTVS S ASTKGP S VFPLAP S SKSTSGGT A ALGCLVKD YFPEP VTVS WN S G ALTS G VHT FP AVLQS SGLYSLS S WTVPS S SLGTQTYICNVNHKP SNTK 10 VDKRVEPKHHHHHH SEQ ID NO: 8 (AM-VH1)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWISWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV 15 S WN S G ALTS GVHTFP AVLQS S GLY S LSS WTVPS S SLGT QT YICN VNHKP SNT
KVDKRVEPKHHHHHH SEQ ID NO: 9 (AM-VH2-E40)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWIEWVRQAPGQGLEWMGRI LPG S G STN Y AQKF QGRVTIT AD ESI ST A YMELS SLRS EDT A V Y Y C TEG YE YDG 20 FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV
SWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55)
Q V Q L VQ S G AE VKKPG S S VKVSCK ATG YTF SR YWIS W VRQ APGQ GLEWMGEI 25 LPGSGSTNYAQKFQGRVT1T ADESTSTAYMELSSLRSEDTAVYY CTEGYEYDG
FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD YFPEP VTV S WN S G ALTS GVHTFP AVLQS SGLY S LS S V VT VP S S S LGTQT YICN VNHKP SNT KVDKRVEPKHHHHHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55) WO 2013/072513 PCT/EP2012/072932 60
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWIEWVRQAPGQGLEWMGEI LP G S G STN Y AQKF QGR VTIT ADES TST A YMELS S LRS EDT A VY Y CTEGYEYDG FD Y W GQGTTVTV S S ASTKGP S VFPLAP S SKST S GGT A ALGCLVKD YFPEP VTV S WNSGALTSGVHTFP AVLQS SGLY S LS S VVTVPS S SLGTQTYICNVNEtKPSNT KVDKRVEPKHHHHHH SEQ ID NO; 12 (AM-VL-C)
DVLLSQTPLSLPVSLGDQATISCRSSQSIVYSNGNTYLEWYLQKPGQSPKLLIY
RVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHIPYTFGGGTK
LEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS
GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF
NRGEC SEQ ID NO: 13 (AM-VL1)
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLNWFQQRPGQSPRRLIY RV SNRD S G VPDRF S GS GS GTDFT LKISRVE AED V GV Y Y CF QGSHIP YTF GQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQ SGNSQES VTEQD SKD STYSLS STLTLSKAD YEKHKVY ACE VTHQGLSSP VTKS FNRGEC SEQ ID NO: 14 (AM-VL2-E40)
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLEWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTV AAP S VFIFPPSDEQLKSGT AS WCLLNNFYPRE AKV Q WKVDN ALQ SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC 13. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to 12 to be used in combination with vasopressors e.g. catecholamine and/ or fluids administered intravenously. 14. ADM antibody or adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease of a patient according to any of the claims 1 to WO 2013/072513 PCT/EP2012/072932 61 13 or a combination according to claim 10 to be used in combination with ADM binding protein and/or further active ingredients. 15. Pharmaceutical formulation comprising an antibody or fragment or scaffold according to any of claims 1 to 14. 16. Pharmaceutical formulation according to claim 15 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 17. Pharmaceutical formulation according to claim 15 wherein said pharmaceutical formulation is in a freeze-dried state. 18. Pharmaceutical formulation according to any of claims 15 to 16, wherein said pharmaceutical formulation is administered intra-muscular. 19. Pharmaceutical formulation according to any of claims 15 to 16, wherein said pharmaceutical formulation is administered intra-vascular. 20. Pharmaceutical formulation according to claim 19, wherein said pharmaceutical formulation is administered via infusion. 21. ADM antibody or an Adrenomedullin antibody fragment or AM non-Ig scaffold, wherein said antibody or fragment or scaffold binds to the N-terminal part (aa 1-21) of Adrenomedullin in, preferably human ADM. 22. Antibody or fragment or scaffold according to claim 2, wherein said antibody or fragment or scaffold recognizes and binds to the N-terminal end (aa 1) of Adrenomedullin.
Further embodiments within the scope of the present invention are set out below: WO 2013/072513 PCT/EP2012/072932 1. 2. 5 3. 4. 10 5. 15 6. 20 7. 8. 25 9. 62
Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient for prevention of organ dysfunction or organ failure. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease according to claim 1 wherein said organ is kidney. ADM antibody or an adrenomedullin antibody fragment according to claim 1 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. ADM antibody or an adrenomedullin antibody fragment according any of claims 1 to 3 wherein said antibody or fragment binds to the N-terminal part (aa 1-21) of adrenomedullin . ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 4, wherein said antibody or fragment recognizes and binds to the N-terminal end (aal) of adrenomedullin. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 5, wherein said antibody or said fragment is an ADM stabilizing antibody or fragment that enhances the tl/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100%. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 6, wherein said antibody blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 7 wherein said disease is selected from the group comprising sepsis, diabetis, cancer, heart failure, and shock. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 8 wherein said patient is an ICU patient. WO 2013/072513 PCT/EP2012/072932 63 10. ADM antibody or an adrenomedullin antibody fragment for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 9 wherein said antibody or fragment is a modulating antibody or fragment that enhances the tl/2 half retention time of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100% and that blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 11. Pharmaceutical formulation comprising an antibody or fragment according to any of claims 1 to 10. 12. Pharmaceutical formulation according to claim 11 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 13. Pharmaceutical formulation according to claim 11 wherein said pharmaceutical formulation is in a freeze-dried state. 14. Pharmaceutical formulation according to any of claims 11 to 12, wherein said pharmaceutical formulation is administered intra-muscular. 15. Pharmaceutical formulation according to any of claims 11 to 12, wherein said pharmaceutical formulation is administered intra-vascular. 16. Pharmaceutical formulation according to claim 15, wherein said pharmaceutical formulation is administered via infusion.
Further embodiments within the scope of the present invention are set out below: 1. Adrenomedullin (ADM) antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold for use in therapy of a chronical or acute disease or acute condition of a patient for prevention or reduction of organ dysfunction or prevention of organ failure in said patient. 2. ADM antibody or an adrenomedullin antibody fragment or ADM non-Ig scaffold for use in therapy of a chronical or acute disease or acute disease according to claim 1 wherein said organ is kidney or liver. WO 2013/072513 PCT/EP2012/072932 64 3. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to claim 1 or 2 wherein said ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold is a non-neutralizing ADM antibody or a nonneutralizing adrenomedullin antibody fragment or a non-neutralizing ADM non-IG scaffold 4. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 or 3 wherein the antibody format is selected from the group comprising Fv fragment, scFv fragment, Fab fragment, scFab fragment, (Fab)2 fragment and scFv-Fc Fusion protein. 5. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according any of claims 1 to 4 wherein said antibody or fragment or scaffold binds to the N-terminal part (aa 1-21) of adrenomedullin . 6. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 5, wherein said antibody or fragment or scaffold recognizes and binds to the N-terminal end (aal) of adrenomedullin. 7. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 6, wherein said antibody or said fragment or scaffold is an ADM stabilizing antibody or fragment or scaffold that enhances the half life (tl/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100%. 8. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold according to any of claims 1 to 7, wherein said antibody or fragment or scaffold blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 9. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronical or acute disease or acute condition of a patient according to any of claims 1 to 8 wherein said disease is selected from the group comprising sepsis, diabetis, cancer, heart failure, and shock. 10. ADM antibody or an adrenomedullin antibody fragment according to any of claims 1 to 9, wherein said antibody or fragment is a human monoclonal antibody or fragment that WO 2013/072513 PCT/EP2012/072932 65 binds to ADM or an antibody fragment thereof wherein the heavy chain comprises the sequences SEQ ID NO: 1
GYTFSRYW
SEQ ID NO: 2 ILPGSGST
SEQ ID NO: 3 TEGYEYDGFDY and wherein the light chain comprises the sequences SEQ ID NO:4
QSIVYSNGNTY
SEQ ID NO: 5 RVS SEQ ID NO: 6 FQGSHIPYT. 11. A human monoclonal antibody or fragment that binds to ADM or an antibody fragment thereof according to claim 10 wherein said antibody or fragment comprises a sequence selected from the group comprising : SEQ ID NO: 7 (AM-VH-C)
QVQLQQSGAELMKPGASVKISCKATGYTFSRYWIEWVKQRPGHGLEWIGEIL P GS GSTN YNEKFKGKATIT ADTS SNT A YMQLS S LTS ED S A V Y Y C TE GYE YDGF WO 2013/072513 PCT/EP2012/072932 66
DYWGQGTTLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVS
WNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTK
VDKRVEPKHHHHHH SEQ ID NO: 8 (AM-VH1)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWISWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FD YW GQGTT VT V S S ASTKGP S VFPLAP S SKSTSGGT AALGCL VKD YFPEP VT V S WN SG ALTSG VHTFP AVLQS SGLY SLS S V VTVPS S SLGTQT YICN VNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 9 (AM-VH2-E40)
QVQLVQSGAEVKKPGSSVKVSCKASGYTFSRYWIEWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCL VKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55)
QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWISWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSG VHTFP AVLQS SGLYSLS SVVTVPSS SLGTQTYICN VNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55)
Q VQLV Q S G AE VKKP GS S VKV S CKATG YTF SRYWIE WVRQAP GQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN VNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 12 (AM-VL-C)
DVLLSQTPLSLPVSLGDQATiSCRSSQSIVYSNGNTYLEWYLQKPGQSPKLLIY
RVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHIPYTFGGGTK WO 2013/072513 PCT/EP2012/072932 67
LEIKRT VA APS VFIFPP SDEQLKSGT AS V VC LLNNF YPREAKV QWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF NRGEC SEQ ID NO: 13 (AM-VL1)
DWMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLNWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTVA AP S VFIFPPSDEQLKSGT AS VVCLLNNF YPREAKV Q WKVDN ALQ SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC SEQ ID NO: 14 (AM-VL2-E40)
DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLEWFQQRPGQSPRRLIY RV SNRD SGVPDRFS GSGSGTDFTLKISRVEAED VG VYY CFQGSHIP YTF GQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQ SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVY ACEVTHQGLSSPVTKS FNRGEC 12. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronical or acute disease of a patient according to any of claims 1 to 11 wherein said antibody or fragment or scaffold is a modulating antibody or fragment or scaffold that enhances the half life ( tl/2 half retention time) of adrenomedullin in serum, blood, plasma at least 10 %, preferably at least 50 %, more preferably >50 %, most preferably >100% and that blocks the bioactivity of ADM to less than 80 %, preferably less than 50%. 13. ADM antibody or an adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease or acute condition of a patient according to any of the claims 1 to 12 to be used in combination with vasopressors e.g. catecholamine and/ or fluids administered intravenously. 14. ADM antibody or adrenomedullin antibody fragment or ADM non-IG scaffold for use in therapy of a chronic or acute disease or acute condition of a patient according to any of the claims 1 to 13 or a combination according to claim 13 to be used in combination with ADM binding protein and/or further active ingredients. WO 2013/072513 PCT/EP2012/072932 68 15. Pharmaceutical formulation comprising an antibody or fragment according to any of claims 1 to 13. 16. Pharmaceutical formulation according to claim 14 wherein said pharmaceutical formulation is a solution, preferably a ready-to-use solution. 17. Pharmaceutical formulation according to claim 14 wherein said pharmaceutical formulation is in a freeze-dried state. 18. Pharmaceutical formulation according to any of claims 14 to 15, wherein said pharmaceutical formulation is administered intra-muscular. 19. Pharmaceutical formulation according to any of claims 14 to 15, wherein said pharmaceutical formulation is administered intra-vascular. 20. Pharmaceutical formulation according to claim 18, wherein said pharmaceutical formulation is administered via infusion. WO 2013/072513 PCT/EP2012/072932 69
EXAMPLES
It should be emphasized that the antibodies, antibody fragments and non-Ig scaffolds of the example portion in accordance with the invention are binding to ADM, and thus should be considered as anti-ADM antibodies/antibody ffagments/non-Ig scaffolds.
Example 1
Generation of Antibodies and determination of their affinity constants
Several human and murine antibodies were produced and their affinity constants were determined (see tables 1 and 2).
Peptides/ conjugates for Immunization:
Peptides for immunization were synthesized, see Table 1, (JPT Technologies, Berlin, Germany) with an additional N-terminal Cystein (if no Cystein is present within the selected ADM-sequence) residue for conjugation of the peptides to Bovine Serum Albumin (BSA). The peptides were covalently linked to BSA by using Sulfolink-coupling gel (Perbio-science, Bonn, Germany). The coupling procedure was performed according to the manual of Perbio.
The murine antibodies were generated according to the following method: A Balb/c mouse was immunized with lOOpg Peptide-BSA-Conjugate at day 0 and 14 (emulsified in ΙΟΟμΙ complete Freund’s adjuvant) and 50pg at day 21 and 28 (in 100μ1 incomplete Freund’s adjuvant). Three days before the fusion experiment was performed, the animal received 50pg of the conjugate dissolved in ΙΟΟμΙ saline, given as one intraperitoneal and one intra-venous injection.
Spenocytes from the immunized mouse and cells of the myeloma cell line SP2/0 were fused with 1ml 50% polyethylene glycol for 30s at 37°C. After washing, the cells were seeded in 96-well cell culture plates. Hybrid clones were selected by growing in HAT medium [RPMI 1640 culture medium supplemented with 20% fetal calf serum and HAT-Supplement]. After two weeks the HAT medium is replaced with HT Medium for three passages followed by returning to the normal cell culture medium. WO 2013/072513 PCT/EP2012/072932 70
The cell culture supernatants were primary screened for antigen specific IgG antibodies three weeks after fusion. The positive tested microcultures were transferred into 24-well plates for propagation. After retesting, the selected cultures were cloned and recloned using the limiting-dilution technique and the isotypes were determined. (see also Lane, R.D. “A short-duration polyethylene glycol fusion technique for increasing production of monoclonal antibody-secreting hybridomas”, J. Immunol. Meth. 81: 223-228; (1985), Ziegler, B. el al “Glutamate decarboxylase (GAD) is not detectable on the surface of rat islet cells examined by cytofluorometry and complement-dependent antibody-mediated cytotoxicity of monoclonal GAD antibodies”, Horm. Metab. Res. 28: 11-15, (1996)).
Mouse monoclonal antibody production:
Antibodies were produced via standard antibody production methods (Marx et al, Monoclonal Antibody Production, ATLA 25, 121, 1997,) and purified via Protein A. The antibody purities were > 95% based on SDS gel electrophoresis analysis.
Human Antibodies
Human Antibodies were produced by means of phage display according to the following procedure:
The human naive antibody gene libraries HAL7/8 were used for the isolation of recombinant single chain F-Variable domains (scFv) against adrenomedullin peptide. The antibody gene libraries were screened with a panning strategy comprising the use of peptides containing a biotin tag linked via two different spacers to the adrenomedullin peptide sequence. A mix of panning rounds using non-specifically bound antigen and streptavidin bound antigen were used to minimize background of non-specific binders. The eluted phages from the third round of panning have been used for the generation of monoclonal scFv expressing E.coli strains. Supernatant from the cultivation of these clonal strains has been directly used for an antigen ELISA testing (see also Hust, M., Meyer, T., Voedisch, B., Rulker, T., Thie, H., El-Ghezal, A., Kirsch, M.I., Schutte, M., Hehnsing, S., Meier, D., Schirrmann, T., Dubel, S., 2011. A human scFv antibody generation pipeline for proteome research. Journal of Biotechnology 152, 159-170; Schutte, M., Thullier, P., Pelat, T., Wezler, X., Rosenstock, P., Hinz, D., Kirsch, M.I.,Hasenberg, M., Frank, R., Schirrmann, T., Gunzer, M., Hust, M., Diibel, S., 2009. Identification of a putative Crf splice variant and generation of recombinant antibodies for the specific detection of Aspergillus fumigatus. PLoS One 4, e6625). WO 2013/072513 PCT/EP2012/072932 71
Positive clones have been selected based on positive ELISA signal for antigen and negative for streptavidin coated micro titer plates. For further characterizations the scFv open reading frame has been cloned into the expression plasmid pOPE107 (Hust et al., J. Biotechn. 2011), captured from the culture supernatant via immobilised metal ion affinity chromatography and 5 purified by a size exclusion chromatography.
Affinity Constants
To determine the affinity of the antibodies to Adrenomedullin, the kinetics of binding of Adrenomedullin to immobilized antibody was determined by means of label-free surface 10 plasmon resonance using a Biacore 2000 system (GE Healthcare Europe GmbH, Freiburg, Germany). Reversible immobilization of the antibodies was performed using an anti-mouse Fc antibody covalently coupled in high density to a CMS sensor surface according to the manufacturer's instructions (mouse antibody capture kit; GE Healthcare). (Lorenz et al.“ Functional Antibodies Targeting IsaA of Staphylococcus aureus Augment Host Immune 15 Response and Open New Perspectives for Antibacterial Therapy44; Antimicrob Agents Chemother. 2011 January; 55(1): 165-173.)
The monoclonal antibodies were raised against the below depicted ADM regions of human and murine ADM, respectively. The following table represents a selection of obtained 20 antibodies used in further experiments. Selection was based on target region:
Table 1:
Sequence Number Antigen/Immunogen ADM Region Designation Affinity constants Kd (M) SEQ ID: 15 YRQSMNNFQGLRSFGCRFGTC 1-21 NT-H 5.9 x 10"y SEQ ID: 16 CTVQKLAHQIYQ 21-32 MR-H 2 x 10^ SEQ ID: 17 C APRSKIS PQGY -NH2 C-42-52 CT-H 1.1 X 10’y SEQ ID: 18 YRQSMNQGSRSNGCRFGTC 1-19 NT-M 3.9 x 10~y SEQ ID: 19 CTFQKLAHQIYQ 19-31 MR-M 4.5 x 10'1U SEQ ID: 20 C APRNKISPQGY -ΝΉ2 C-40-50 CT-M 9 x 10'y WO 2013/072513 PCT/EP2012/072932 72
The following is a list of further obtained monoclonal antibodies: List of anti-ADM-antibodies
Table 2:
Target Source Klone number Affinity | (M) max inhibition bioassay (%) (see example 2) NT-M Mouse ADM/63 5.8xlO'v 45 Mouse ADM/364 2.2x10'** 48 Mouse ADM/365 3.0x10'** Mouse ADM/366 1.7x10'** ! Mouse ADM/367 1.3x10'** Mouse ADM/368 1.9 xlO'** Mouse ADM/369 2.0x10^ Mouse ADM/370 1.6x10'“ Mouse ADM/371 2.0x10^ Mouse ADM/372 ~2.5xl0'** ' Mouse ADM/373 1.8x10^ Mouse ADM/377 1.5 xl0'“ Mouse ADM/378 2.2x10'“ Mouse ADM/379 1.6x10'“ Mouse ADM/380 1.8 xltP Mouse ADM/381 2.4x10'“ Mouse ADM/382 1.6x10'“ Mouse ADM/383 1.8x10'“ Mouse ADM/384 1.7x10'“ Mouse ADM/385 1.7x10'“ Mouse ADM/403 1.2x10'“ Mouse ADM/395 1.2x10'“ Mouse ADM/396 3.0 x 10* Mouse ADM/397 1.5x10'“ MR-M Mouse ADM/38 4.5x1 O'10 68 MR-M Mouse ADM/39 S.OxlfT5 72 PCT/EP2012/072932 WO 2013/072513 73 CT-M Mouse 1 ADM/65 9.0x10'y © o CT-M Mouse ADM/66 1.6xl0“s 100 NT-H Mouse ADM/33 5.9xl0*8 1 38 NT-H Mouse ADM/34 1.6xl0"s 22 MR-H Mouse ADM/41 1.2x1 O'3 67 MR-H Mouse ADM/42 <lxl0's MR-H Mouse ADM/43 2.0x10'y 73 MR-H Mouse ADM/44 <1x10‘s CT-H Mouse ADM/15 <lxl0‘s CT-H Mouse ADM/16 l.lxl0'y 100 CT-H Mouse ADM/17 3.7xl0'y 100 CT-H Mouse ADM/18 <lxl0"8 hADM Phage display ADM/A7 <lxl0‘M Phage display ADM/B7 <lxl0~B Phage display ADM/C7 <lxl0's Phage display ADM/G3 <1x10s Phage display ADM/B6 <lxl0's Phage display ADM/B11 <1x10‘8 Phage display ADM/D8 <1x10^ Phage display ADM/D11 <1x10'* Phage display ADM/G12 <1x10'8
Generation of antibody fragments by enzymatic digestion:
The generation of Fab and F(ab)2 fragments was done by enzymatic digestion of the murine full length antibody NT-M. Antibody NT-M was digested using a) the pepsin-based F(ab)2 Preparation Kit (Pierce 44988) and b) the papain-based Fab Preparation Kit (Pierce 44985). The fragmentation procedures were performed according to the instructions provided by the supplier. Digestion was carried out in case of F(ab)2-fragmentation for 8h at 37°C. The Fab-fragmentation digestion was carried out for 16h, respectively. WO 2013/072513 PCT/EP2012/072932 74
Procedure for Fab Generation and Purification:
The immobilized papain was equilibrated by washing the resin with 0.5 ml of Digestion Buffer and centrifuging the column at 5000 x g for 1 minute. The buffer was discarded afterwards. The desalting column was prepared by removing the storage solution and washing it with digestion buffer, centrifuging it each time afterwards at 1000 x g for 2 minutes. 0.5ml of the prepared IgG sample where added to the spin column tube containing the equilibrated Immobilized Papain. Incubation time of the digestion reaction was done for 16h on a tabletop rocker at 37°C. The column was centrifuged at 5000 x g for 1 minute to separate digest from the Immobilized Papain. Afterwards the resin was washed with 0.5ml PBS and centrifuged at 5000 x g for 1 minute. The wash fraction was added to the digested antibody that the total sample volume was 1.0ml. The NAb Protein A Column was equilibrated with PBS and IgG Elution Buffer at room temperature. The column was centrifuged for 1 minute to remove storage solution (contains 0.02% sodium azide) and equilibrated by adding 2ml of PBS, centrifuge again for 1 minute and the flow-through discarded. The sample was applied to the column and resuspended by inversion. Incubation was done at room temperature with end-over-end mixing for 10 minutes. The column was centrifuged for 1 minute, saving the flowthrough with the Fab fragments. (References: Coulter, A. and Harris, R. (1983). J. Immunol. Meth. 59, 199-203.; Lindner I. et al. (2010) {alpha}2-Macroglobulin inhibits the malignant properties of astrocytoma cells by impeding {beta}-catenin signaling. Cancer Res. 70, 277-87.; Kaufinann B. et al. (2010) Neutralization of West Nile virus by cross-linking of its surface proteins with Fab fragments of the human monoclonal antibody CR4354. PNAS. 107, 18950-5.; Chen X. et al (2010) Requirement of open headpiece conformation for activation of leukocyte integrin αχβ2. PNAS. 107, 14727-32.; Uysal H. et al. (2009) Structure and pathogenicity of antibodies specific for citrullinated collagen type II in experimental arthitis. J. Exp. Med. 206, 449-62.; Thomas G. M. et al. (2009) Cancer cell-derived microparticles bearing P-selectin glycoprotein ligand 1 accelerate thrombus formation in vivo. J. Exp. Med. 206, 1913-27.; Kong F. et al. (2009) Demonstration of catch bonds between an integrin and its ligand. J. Cell Biol. 185,1275-84.)
Procedure for generation and purification of F(ab'H Fragments:
The immobilized Pepsin was equilibrated by washing the resin with 0.5 ml of Digestion Buffer and centrifuging the column at 5000 x g for 1 minute. The buffer was discarded afterwards. The desalting column was prepared by removing the storage solution and washing WO 2013/072513 PCT/EP2012/072932 75 it with digestion buffer, centrifuging it each time afterwards at 1000 x g for 2 minutes. 0.5ml of the prepared IgG sample where added to the spin column tube containing the equilibrated Immobilized Pepsin. Incubation time of the digestion reaction was done for 16h on a tabletop rocker at 37°C. The column was centrifuged at 5000 χ g for 1 minute to separate digest from the Immobilized Papain. Afterwards the resin was washed with 0.5mL PBS and centrifuged at 5000 χ g for 1 minute. The wash fraction was added to the digested antibody that the total sample volume was 1.0ml. The NAb Protein A Column was equilibrated with PBS and IgG Elution Buffer at room temperature. The column was centrifuged for 1 minute to remove storage solution (contains 0.02% sodium azide) and equilibrated by adding 2mL of PBS, centrifuge again for 1 minute and the flow-through discarded. The sample was applied to the column and resuspended by inversion. Incubation was done at room temperature with end-over-end mixing for 10 minutes. The column was centrifuged for 1 minute, saving the flowthrough with the Fab fragments. (References: Mariani, M., el al. (1991). A new enzymatic method to obtain high-yield F(ab')2 suitable for clinical use from mouse IgGl. Mol.Immunol. 28: 69-77.;Beale, D. (1987). Molecular fragmentation: Some applications in immunology. Exp Comp Immunol 11:287-96.; Ellerson, J.R., et al. (1972). A fragment corresponding to the CH2 region of immunoglobulin G (IgG) with complement fixing activity. FEBS Letters 24(3):318-22.; Kerbel, R.S. and Elliot, B.E. (1983). Detection of Fc receptors. Meth Enzymol 93:113-147.; Kulkami, P.N., et al. (1985). Conjugation of methotrexate to IgG antibodies and their F(ab')2 fragments and the effect of conjugated methotrexate on tumor growth in vivo. Cancer Immunol Immunotherapy 19:211-4.; Lamoyi, E. (1986). Preparation of F(ab')2 Fragments from mouse IgG of various subclasses. Meth Enzymol 121:652-663.; Parham, P., et al. (1982), Monoclonal antibodies: purification, fragmentation and application to structural and functional studies of class I MHC antigens. J Immunol Meth 53:133-73.; Raychaudhuri, G., et al. (1985). Human IgGl and its Fc fragment bind with different affinities to the Fc receptors on the human U937, HL-6G and ML-1 cell lines. Mol Immunol 22(9):1009-19.; Rousseaux, J., et al. (1980). The differential enzyme sensitivity of rat immunoglobulin G subclasses to papain an pepsin. Mol Immunol 17:469-82.; Rousseaux, J., et al. (1983). Optimal condition for the preparation of Fab and F(ab')2 fragments from monoclonal IgG of different rat IgG subclasses. J Immunol Meth 64:141-6.; Wilson, K.M., et al. (1991). Rapid whole blood assay for HIV-1 seropositivity using an Fab-peptide conjugate, J Immunol Meth 138:111-9.) WO 2013/072513 PCT/EP2012/072932 76 NT-H-Antibody Fragment Humanization
The antibody fragment was humanized by the CDR-grafting method (Jones, P. T., Dear, P. H., Foote, J., Neuberger, M. S., and Winter, G. (1986) Replacing the complementaritydetermining regions in a human antibody with those from a mouse. Nature 321, 522-525). 5 The following steps where done to achieve the humanized sequence:
Total RNA extraction: Total RNA was extracted from NT-H hybridomas using the Qiagen kit.
First-round RT-PCR: QIAGEN® OneStep RT-PCR Kit (Cat No. 210210) was used. RT-PCR was performed with primer sets specific for the heavy and light chains. For each RNA 10 sample, 12 individual heavy chain and 11 light chain RT-PCR reactions were set up using degenerate forward primer mixtures covering the leader sequences of variable regions. Reverse primers are located in the constant regions of heavy and light chains. No restriction sites were engineered into the primers.
Reaction Setup: 5x QIAGEN® OneStep RT-PCR Buffer 5.0 μΐ, dNTP Mix (containing 10 15 mM of each dNTP) 0.8 μΐ, Primer set 0.5 μΐ, QIAGEN® OneStep RT-PCR Enzyme Mix 0.8 μΐ, Template RNA 2.0 μΐ, RNase-free water to 20.0 μΐ, Total volume 20.0 μΐ PCR condition: Reverse transcription: 50°C, 30 min; Initial PCR activation: 95°C, 15 min
Cycling: 20 cycles of 94°C, 25 sec; 54°C, 30 sec; 72°C, 30 sec; Final extension: 72°C, 10 min
Second-round semi-nested PCR: The RT-PCR products from the first-round reactions were 20 further amplified in the second-round PCR. 12 individual heavy chain and 11 light chain RT-PCR reactions were set up using semi-nested primer sets specific for antibody variable regions.
Reaction Setup: 2x PCR mix 10 μΐ; Primer set 2 μΐ; First-round PCR product 8 μΐ; Total volume 20 μΐ; Hybridoma Antibody Cloning Report 25 PCR condition: Initial denaturing of 5 min at 95°C; 25 cycles of 95°C for 25 sec, 57°C for 30 sec, 68°C for 30 sec; Final extension is 10 min 68°C. WO 2013/072513 PCT/EP2012/072932 77
After PCR is finished, run PCR reaction samples onto agarose gel to visualize DNA fragments amplified.After sequencing more than 15 cloned DNA fragments amplified by nested RT-PCR, several mouse antibody heavy and light chains have been cloned and appear correct. Protein sequence alignment and CDR analysis identifies one heavy chain and one light chain. After alignment with homologous human framework sequences the resulting humanized sequence for the variable heavy chain is the following: see figure 6 (As the amino acids on positions 26, 40 and 55 in the variable heavy chain and amino acid on position 40 in the variable light are critical to the binding properties, they may be reverted to the murine original. The resulting candidates are depicted below) (Padlan, E. A. (1991) A possible procedure for reducing the immunogenicity of antibody variable domains while preserving their ligand-binding properties. Mol. Immunol. 28, 489-498.; Harris, L. and Bajorath, J. (1995) Profiles for the analysis of immunoglobulin sequences: Comparison of V gene subgroups. Protein Sci. 4, 306-310.).
Annotation for the antibody fragment sequences (SEQ ID NO: 7-14): bold and underline are the CDR 1, 2, 3 in chronologically arranged; italic are constant regions; hinge regions are highlighted with bold letters and the histidine tag with bold and italic letters; framework point mutation have a grey letter-background. SEQ ID NO: 7 (AM-VH-C)
OVOLOOSGAELMKPGASVKISCKATGYTFSRYW1EWVKORPGHGLEWIGEILPGSG STN YNEKFKGKATIT AD T S SNTA YMQLSSLTSEDSA VYYCTEGYEYDGFDYWGOGTTLT VSSASTKGPSVFPLAPSSKSTSGGTAALGCL VKD YFPEPVTVSWNSGAL TSG VHTFPA VLQS SGLYSLSSWTVPSSSL GTQTY1CNVNHKPSNTK VDKR VEY'KMHHHHH SEQ ID NO: 8 (AM-VH1)
OVOLVOSGAEVKKPGSSVKVSCKASGYTFSRYWISWVROAPGOGLEWMGRILPGS GSTNY AOKFOGRVT ΙΤΑΡΕ5ΤΛ7>ί YMELSSLRSEDTA VYYCTEGYEYDGFDYWGOG TTV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKRVYYKHHHHHH SEQ ID NO: 9 (AM-VH2-E40)
OVOLVOSGAEVKKP GS S VKVSCKAS GYTFSRYWlBWVROAPGOGLEWMGRlLPGS GSTNY AOKF QGRVTIT ADESTSTA YMELSSLRSEDTA VYYCTEGYEYDGFDYWGOGTTV
TVSSASTKGPSVFPLAPSSKSTSGGTAALGCL VKDYFPEP VTVSWNSGAL TSG VHTFPA VLQ SSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEYKHHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55) WO 2013/072513 PCT/EP2012/072932 78 ovolvqsgaevkkpgssvkvsckaIgytfsrywiswvrqapgoglewmgIilpgs
GSTNYAOKFQGRVTITADEYrm YMELSSLRSEDTA VYYCTEGYEYDGFDYWGOGTTV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSSSLGTQTYICN VNHKPSNTKVDKR VEFKHHHIIHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55)
OVOLVOSGAEVKKPGSSVKVSCKAiGYTFSRYWllWVRQAPGOGLEWMGilLPGS GSTNYAQKFQGRVTITADE5,AS'7M YMELSSLRSEDTA VYYCTEGYEYDGFDYWGOGTTV TVSSASTKGPSVFPLAPSSKSTSGGTAALGCL VKD YFPEP VTVSWNSGAL TSG VHTFPA VLQ SSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNTKVDKRVYYKHHHHHH SEQ ID NO: 12 (AM-VL-C)
DVLLSOTPLSLPVSLGDOATISCRSSOSIVYSNGNTYLEWYLOKPGOSPKLLIYRVSN
RFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYYCFQGSHIPYTFGGGTKLEIKRTVA
APSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS
KDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC SEQ ID NO: 13 (AM-VLl)
DWMTOSPLSLPVTLGOPASISCRSSOSIVYSNGNTYLNWFOORPGOSPRRLIYRVSN
RDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFOGSHIPYTFGOGTKLEIKRTVA
APSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS
KDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC SEQ ID NO: 14 (AM-VL2-E40)
RDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFOGSHIPYTFGOGTKLEIKRTVA APSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS KD STY S LS STLTLSKAD YEKHKVY ACE VTHQGLS SP VTKS FNRGEC
Example 2
Effect of selected anti-ADM-antibodies on anti-ADM-bioactivity
The effect of selected ADM-antibodies on ADM-bioactivity was tested in an human recombinant Adrenomedullin receptor cAMP functional assay (Adrenomedullin Bioassay).
Testing of antibodies targeting human or mouse adrenomedullin in human recombinant Adrenomedullin receptor cAMP functional assay (Adrenomedullin Bioassay) WO 2013/072513 PCT/EP2012/072932 79
Materials:
Cell line: CHO-K1
Receptor: Adrenomedullin (CRLR + RAMP3)
Receptor Accession Number Cell line: CRLR: U17473; RAMP3: AJ001016 CHO-K1 cells expressing human recombinant adrenomedullin receptor (FAST-027C) grown prior to the test in media without antibiotic were detached by gentle flushing with PBS-EDTA (5 mM EDTA), recovered by centrifugation and resuspended in assay buffer (KRH: 5 mM KC1, 1.25 mM MgS04, 124 mM NaCl, 25 mM HEPES, 13.3 mM Glucose, 1.25 mM KH2P04, 1.45 mM CaC12, 0.5 g/1 BSA).
Dose response curves were performed in parallel with the reference agonists (hADM or mADM).
Antagonist test (96well):
For antagonist testing, 6 μΐ of the reference agonist (human (5,63nM) or mouse (0,67nM) adrenomedullin) was mixed with 6 μΐ of the test samples at different antagonist dilutions; or with 6 μΐ buffer. After incubation for 60 min at room temperature, 12 μΐ of cells (2,500 cells/well) were added. The plates were incubated for 30 min at room temperature. After addition of the lysis buffer, percentage of DeltaF will be estimated, according to the manufacturer specification, with the FITRF kit from Cis-Bio International (cat n°62AM2 PEB). hADM 22-52 was used as reference antagonist.
Antibodies testing cAMP-HTRF assay
The anti-h-ADM antibodies (NT-H, MR-H, CT-H) were tested for antagonist activity in human recombinant adrenomedullin receptor (FAST-027C) cAMP functional assay in the presence of 5.63nM Human ADM 1-52, at the following final antibody concentrations: 100pg/ml, 20pg/ml, 4μg/ml, 0.8pg/ml, 0.16pg/ml.
The anti-m-ADM antibodies (NT-M, MR-M, CT-M) were tested for antagonist activity in human recombinant adrenomedullin receptor (FAST-027C) cAMP functional assay in the presence of 0.67nM Mouse ADM 1-50, at the following final antibody concentrations: 100pg/ml, 20pg/ml, 4pg/ml, 0.8pg/ml, 0.16pg/ml. Data were plotted relative inhibition vs. antagonist concentration (see figs. 3a to 31). The maximal inhibition by the individual antibody is given in table 3. WO 2013/072513 PCT/EP2012/072932 80
Table 3:
Antibody Maximal inhibition of ADM bioactivity (ADM-Bioassay) (%) NT-H 38 MR-H 73 CT-H 100 NT-M FAB 26 NT-M FAB2 28 NT-M 45 MR-M 66 CT-M 100 Non specific mouse IgG 0
Example 3 5 Data for stabilization of hADM by the anti-ADM antibody
The stabilizing effect of human ADM by human ADM antibodies was tested using a hADM immunoassay. 10 Immunoassay for the quantification of human Adrenomedullin
The technology used was a sandwich coated tube luminescence immunoassay, based on Acridinium ester labelling. 15 Labelled compound (tracer): 100pg (lOOul) CT-H (lmg/ ml in PBS, pH 7.4, AdrenoMed AGGermany) was mixed with 10μ1 Acridinium NHS-ester (lmg/ ml in acetonitrile, InVent GmbH, Germany) (EP 0353971) and incubated for 20min at room temperature. Labelled CT-H was purified by Gel-filtration HPLC on Bio-Sil® SEC 400-5 (Bio-Rad Laboratories, Inc., USA) The purified CT-H was diluted in (300 mmol/L potassiumphosphate, 100 mmol/L 20 NaCl, 10 mmol/L Na-EDTA, 5 g/L Bovine Serum Albumin, pH 7.0). The final concentration was approx. 800.000 relative light units (RLU) of labelled compound (approx. 20ng labeled antibody) per 200 pL. Acridinium ester chemiluminescence was measured by using an AutoLumat LB 953 (Berthold Technologies GmbH &amp; Co. KG). PCT/EP2012/072932 WO 2013/072513 81
Solid phase: Polystyrene tubes (Greiner Bio-One International AG, Austria) were coated (18h at room temperature) with MR-H (AdrenoMed AG, Germany) (1.5 μg MR-H/0.3 mL 100 mmol/L NaCl, 50 mmol/L TRIS/HC1, pH 7.8). After blocking with 5% bovine serum albumine, the tubes were washed with PBS, pH 7.4 and vacuum dried.
Calibration:
The assay was calibrated, using dilutions of hADM (BACHEM AG, Switzerland) in 250 mmol/L NaCl, 2 g/L Triton X-100, 50 g/L Bovine Serum Albumin, 20 tabs/L Protease Inhibitor Cocktail (Roche Diagnostics AG, Switzerland)) hADM Immunoassay: 50 μΐ of sample (or calibrator) was pipetted into coated tubes, after adding labeleld CT-H (200μ1), the tubes were incubated for 4h at 4°C. Unbound tracer was removed by washing 5 times (each 1ml) with washing solution (20mM PBS, pH 7.4, 0.1 % Triton X-100).
Tube-bound chemiluminescence was measured by using the LB 953
Figure 4 shows a typical hADM dose/ signal curve. And an hADM dose signal curve in the presence of 100 pg/niL antibody NT-H. NT-H did not affect the described hADM immunoassay.
Stability of human Adrenomedullin:
Human ADM was diluted in human Citrate plasma (final concentration IGnM) and incubated at 24 °C. At selected time points, the degradation of hADM was stopped by freezing at -20 °C. The incubation was performed in absence and presence of NT-H (100pg/ml). The remaining hADM was quantified by using the hADM immunoassay described above.
Figure 5 shows the stability of hADM in human plasma (citrate) in absence and in the presence of NT-H antibody. The half life of hADM alone was 7,8h and in the presence of NT-H, the half life was I8,3h. (2.3 times higher stability). WO 2013/072513 PCT/EP2012/072932 82
Example 4
Sepsis Mortality (early treatment)
Animal model 12-15 week old male C57B1/6 mice (Charles River Laboratories, Germany) were used for the study. Peritonitis had been surgically induced under light isofluran anesthesia. Incisions were made into the left upper quadrant of the peritoneal cavity (normal location of the cecum). The cecum was exposed and a tight ligature was placed around the cecum with sutures distal to the insertion of the small bowel. One puncture wound was made with a 24-gauge needle into the cecum and small amounts of cecal contents were expressed through the wound. The cecum was replaced into the peritoneal cavity and the laparotomy site was closed. Finally, animals were returned to their cages with free access to food and water. 500μ1 saline were given s.c. as fluid replacement.
Application and dosage of the compound (NT-M, MR-M, CT-M)
Mice were treated immediately after CLP (early treatment). CLP is the abbreviation for cecal ligation and puncture (CLP).
Study groups
Three compounds were tested versus: vehicle and versus control compound treatment. Each group contained 5 mice for blood drawing after 1 day for BUN (serum blood urea nitrogen test) determination. Ten further mice per each group were followed over a period of 4 days. Group Treatment (10μ1/ g bodyweight) dose/ Follow-Up: 1 NT-M, 0.2 mg/ml survival over 4 days 2 MR-M, 0.2 mg/ml survival over 4 days 3 CT-M, 0.2 mg/ml survival over 4 days 4 non-specific mouse IgG, 0.2 mg/ml survival over 4 days 5 control - PBS ΙΟμΙ/g bodyweight survival over 4 days WO 2013/072513 PCT/EP2012/072932 83
Clinical chemistry
Blood urea nitrogen (BUN) concentrations for renal function were measured baseline and day 1 after CLP. Blood samples were obtained from the cavernous sinus with a capillary under light ether anaesthesia. Measurements were performed by using an AU 400 Olympus 5 Multianalyser. The 4-day mortality is given in table 4. The average BUN concentrations are given in table 5.
Table 4: 4 day mortality survival (%) PBS 0 non-specific mouse IgG 0 CT-M 10 MR-M 30 NT-M 70 Table 5:
Average from 5 animals BUN pre CLP (mM) BUN day 1 (mM) PBS 8.0 23.2 non-specific mouse IgG 7.9 15.5 CT-M 7.8 13.5 MR-M 8.1 24.9 NT-M 8.8 8.2
It can be seen from Table 4 that the NT-M antibody reduced mortality considerably. After 4 days 70 % of the mice survived when treated with NT-M antibody. When treated with MR-M antibody 30 % of the animals survived and when treated with CT-M antibody 10 % of the 15 animals survived after 4 days. In contrast thereto all mice were dead after 4 days when treated with unspecific mouse IgG. The same result was obtained in the control group where PBS (phosphate buffered saline) was administered to mice.
The blood urea nitrogen or BUN test is used to evaluate kidney function, to help diagnose kidney disease, and to monitor patients with acute or chronic kidney dysfunction or failure. 20 The results of the S-BUN Test revealed that the NT-M antibody was the most effective to protect the kidney. WO 2013/072513 PCT/EP2012/072932 84
Sepsis Mortality (late treatment)
Animal model 12-15 week old male C57B1/6 mice (Charles River Laboratories, Germany) were used for the study. Peritonitis had been surgically induced under light isofluran anesthesia. Incisions were made into the left upper quadrant of the peritoneal cavity (normal location of the cecum), The cecum was exposed and a tight ligature was placed around the cecum with sutures distal to the insertion of the small bowel. One puncture wound was made with a 24-gauge needle into the cecum and small amounts of cecal contents were expressed through the wound. The cecum was replaced into the peritoneal cavity and the laparotomy site was closed. Finally, animals were returned to their cages with free access to food and water. 500μ1 saline were given s.c. as fluid replacement.
Application and dosage of the compound (NT-M FAB2) NT-M FAB2 was tested versus: vehicle and versus control compound treatment. Treatment was performed after full development of sepsis, 6 hours after CLP (late treatment). Each group contained 4 mice and were followed over a period of 4 days.
Group Treatment (10μ1/ g bodyweight) dose/ Follow-Up:
Study groups 1 NT-M, FAB2 0.2 mg/ml survival over 4 days 2 control: non-specific mouse IgG, 0.2 mg/ml survival over 4 days 3 vehicle: - PBS ΙΟμΙ/g bodyweight survival over 4 days
Table 6: 4 day mortality survival (%) PBS 0 Non-specific mouse IgG 0 NT-M FAB2 75
It can be seen from Table 6 that the NT-M FAB 2 antibody reduced mortality considerably. After 4 days 75 % of the mice survived when treated with NT-M FAB 2 antibody. In contrast WO 2013/072513 PCT/EP2012/072932 85 thereto all mice were dead after 4 days when treated with non-specific mouse IgG. The same result was obtained in the control group where PBS (phosphate buffered saline) was administered to mice.
Example 5
Incremental effect of anti-ADM antibody in CLP-animals on top of antibiotic treatment and circulation stabilization via catecholamines as well as regulation of fluid balance.
Animal model
In this study male C57B1/6 mice (8-12 weeks, 22-30g) were utilized. A polymicrobial sepsis induced by cecal ligation and puncture (CLP) was used as the model for studying septic shock ((Albuszies G, et ah Effect of increased cardiac output on hepatic and intestinal micro circulatory blood flow, oxygenation, and metabolism in hyperdynamic murine septic shock. Crit Care Med 2005;33:2332-8), (Albuszies G, et ah The effect of iNOS deletion on hepatic gluconeogenesis in hyperdynamic murine septic shock. Intensive Care Med 2007;33:1094-101), (Barth E, et ah Role of iNOS in the reduced responsiveness of the myocardium to catecholamines in a hyperdynamic, murine model of septic shock. Crit Care Med 2006;34:307-13), (Baumgart K, et ah Effect of SOD-1 over-expression on myocardial function during resuscitated murine septic shock. Intensive Care Med 2009;35:344-9), (Baumgart K, et ah Cardiac and metabolic effects of hypothermia and inhaled H2S in anesthetized and ventilated mice. Crit Care Med 2010;38:588-95), (Simkova V, et ah The effect of SOD-1 over-expression on hepatic gluconeogenesis and whole-body glucose oxidation during resuscitated, normotensive murine septic shock. Shock 2008;30:578-84), (Wagner F, et ah: Inflammatory effects of hypothermia and inhaled H2S during resuscitated, hyperdynamic murine septic shock. Shock, im Druck), (Wagner F, et ah Effects of intravenous H2S after murine blunt chest trauma: a prospective, randomized controlled trial. Crit Care 2011, submittes for publication)).
After weighing, mice were anesthetized by intraperitoneal injection of 120 pg/g Ketamin, 1.25 pg/g Midazolam and 0.25 pg/g Fentanyl. During the surgical procedure, body temperature was kept at 37-38°C. A 1cm midline abdominal section was performed to get access to the cecum. The cecum then was ligated with 3-0 silk tie close to the basis and a single puncture with a 18-gauge needle was applied. The cecum was returned and the incision WO 2013/072513 PCT/EP2012/072932 86 was closed again (4-0 tie). For the compensation of perioperative loss of liquids, 0.5 ml lacted Ringer’s solution with lpg/g Buprenorphin as analgetic was injected subcutaneously in dorsal dermis. For antibiosis the mice received Ceftriaxon 30pg/g and Clindamycin 30pg/g subcutaneously via the lower extremities.
After CLP surgery the animal were kept in an adequately heated environment with water and food ad libitum.
The covering of liquid requirements were ensured by a dorsal subcutaneous injections with 0.5 ml lactated ringer’s solution with 4 pg/g glucose and Buprenorphin 1 pg/g, which were applied in an 8 hour cycle, after short term anesthesia by isofluran. In addition, antibiosis was maintained by subcutaneous injections of Ceftriaxon 30pg/g and Clindamycin 30pg/g via the lower extremities.
Dosing of test substances
Early treatment
Immediately after the CLP surgery and closing of the incision, the test substance antibody NT-M was applied in a concentration of 500 pg/ml in phosphate buffered saline (PBS) via injection into the penis vein for a dose of 2mg per kg body weight (dose volume 88-120 pi) (5 animals).
Late treatment
After full Sepsis development, 15.5h after CLP surgery, animals were anesthetized as described above and NT-M was applied in a concentration of 500 pg/ml in phosphate buffered saline (PBS) via injection into the penis vein for a dose of 2mg per kg body weight (dose volume 88-120 pi) (3 animals).
The control group (6 animals) received a corresponding amount of the vehicle PBS solution without antibody (4pl/g, 88-120 pi) immediately after CLP surgery.
Study groups and experimental setting
Murine septic shock model under intensive care monitoring: WO 2013/072513 PCT/EP2012/072932 87
Three groups with 3, 5 and 6 animals were monitored. Group 1 (5 animals) received the antibody NT-M 15.5h after CLP, group 2 received the antibody NT-M immediately after CLP surgery and group 3 received a comparable amount of PBS (4pl/g). 16 hour incubation post CLP (to allow the polymicrobial sepsis to progress), the experiment was continued with monitoring and interventions comparable to an intensive medical care regime. Therefore, after weighing the animals were anesthetized as described in the CLP surgery part (except the late treated animals, which were anesthized before treatment). Body temperature was maintained at 37-38°C for the rest of the experiment. After a tracheotomy and intubation, respiration was monitored and supported by laboratory animal lung ventilator Flexivent®, (Emka Technologies, Fi02 0,5, PEEP 10 H20, VT 8pl/g, I:E 1:1,5, AF 70-140 depending on temperature).
Anesthesia was maintained throughout the experiment via the cannulated vena jugularis externa dextra with a continuous infusion of Ketamin 30 pg/gxh and Fentanyl 0.3 pg/gxh. Furthermore, the right aorta carotis communis was cannulated for continuous monitoring of heart rate and the mean arterial pressure (MAP). The mean arterial pressure was maintained at MAP > 65 mmHg via intravenous (V. jugularis) infusion of colloids (80 pL/gxh, Hextend®) and, if needed, Noradrenalin dissolved in colloids as vasopressor. Blood samples (120 μΐ) were taken via the cannulated A. carotis at 0 and 4 hours for determination of creatinine. The bladder was punctured and urine was collected via a bladder catheter. The experiment was either terminated after 6 hours or prior to this, if the MAP > 65 mmHg (V. jugularis) could not be maintained with the vasorpressor dosing.
Measured parameters
The following parameters were measured and analyzed: Total consumption of noradrenalin (pg NA/g), consumption rate of noradrenalin (pg NA/g/h), total volume of urine collected during the experiment, creatinine concentration (pg/mL) at the end of the experiment and mean creatinine clearance (pL/min). WO 2013/072513 Table 7: 88 PCT/EP2012/072932 Total consumption of Noradrenalin (pg NA/g) (Average) consumption rate of Noradrenalin (pg NA/g/h) (Average) Control (mouse IgG) (N—6) 0.17 pg/g 0.032 pg/h/g NT-M (N=5) early treatment 0.07 pg/g 0.012pg/h/g Relative change (early treatment, 59% 62.5% amelioration) (59%) (62.5%) NT-M (N=3) late treatment 0.04 pg/g 0.0075 pg/h/g Relative change (late treatment, 76,5% 76,5% amelioration) (76.5%) (76.5%)
The catecholamine requirement was measured after administration of either non specific mouse IgG to a total of 6 mice as control group, NT-murine antibody to a group of 5 mice 5 immediately after CLP (early treatment) or NT-murine antibody to a group of 3 mice 15.5h after CLP (late treatment).
The reduction of the catecholamine requirement is a measure for the stabilization of the circulation. Thus, the data show that the ADM antibody, especially the NT-M antibody, leads to a considerable stabilization of the circulation and to a considerable reduction of the 10 catecholamine requirement. The circulation-stabilizing effect was given in early treatment (immediately after CLP) and treatment after full sepsis development (late treatment) (see fig. 7).
Regulation of Fluid Balance 15
More positive fluid balance both early in resuscitation and cumulatively over 4 days is associated with an increased risk of mortality in septic shock. The control of the liquid balance is of utmost importance for the course of disease of patients having sepsis, (s. Boyd et al, 2011). Controlling the liquid balance of critical ill patients remains as a substantial 20 challenge in intensive care medicine. As can be seen in table 8 treatment of mice after CLP (experimental procedures see ‘‘Animal Model”) with NT-M antibody lead to an enhancement of the total volume of urine excreted. The urine secreted was approx, three times higher in NT-M-treated animals compared to non-treated mice. The positive treatment effect was given in early- and in late treatment. The fluid balance was improved by about 20-30%, also in both, WO 2013/072513 PCT/EP2012/072932 89 early and late treatment. Thus, the data show that the use of ADM antibody, especially the use of NT ADM antibody, is favorable for regulating the fluid balance in patients, (see table 8 and figures 8 and 9). 5 Table 8
Urine average volume/ g body weight Fluid balance average volume/ g body weight Control (mouse IgG) (N=6) 0.042 ml/g 0,23 ml/g NT-M early (N=5) 0.12 ml 0,18 ml/g Relative change early treatment + 186% -21.7%% NT-M late (N=3) 0.125 ml 0,16 ml/g Relative change late treatment + 198% -30,5%
Improvement of kidney function 10 The combination of acute renal failure and sepsis is associated with a 70 percent mortality, as compared with a 45 percent mortality among patients with acute renal failure alone. (Schrier and Wang, “Mechanisms of Disease Acute Renal Failure and Sepsis”; The New England Journal of Medicine; 351:159-69; 2004). Creatinine concentration and creatinine clearance are standard laboratory parameters for monitoring 15 kidney (dys)function (Jacob, “Acute Renal Failure”, Indian J. Anaesth.; 47 (5): 367-372; 2003). Creatinine and creatinine clearance data from above described animal experiment (early treatment) are given in Table 9. 20 WO 2013/072513 PCT/EP2012/072932 90
Table 9
Kidney function: creatinine concentration (pg/mL) mean creatinine clearance (pL/min) control mouse IgG (MW) 2.6 pg/ml 174 pl/min NT-M (MW) 1.5 pg/ml 373 pl/min Relative change -42% +114% (amelioration) (42%) (114%) 5 In comparision to control septic animals, the creatinine concentration was lowered by 42% and the creatinine clearance was improved by more than 100% as a result of NT-M treatment (Table 9). The data show that the administration of ADM-antibody, especially NT-M, leads to an improvement of kidney function. 10 Improvement of liver inflammatory status
Liver tissue for control and early treated animals was homogenized and lysed in lysing buffer. For cell extract preparation, cells were resuspended, lysed on ice, and centrifuged. The supernatant (protein extract) was stored at -80 °C, Activation of nuclear factor kappa-light-15 chain gene enhancer in B cells (NF-κΒ) was determined as previously described using an electrophoretic mobility shift assay (EMSA)1,2. Cell extracts (1 Opg) were incubated on ice with poly-doxy-inosinic-deoxy-cytidylic acid (poly-dl-dC) and 32P-labeled double stranded oligonucleotide (Biomers, Ulm, Germany) containing the NF-κΒ (HIV KBsite) ( 5’-GGATCCTCAACAGAGGGGACTTTCCGAGGCCA-3’)· Complexes were separated in 20 native polyacrylamide gels, dried and exposed to X-ray films. A phosphorimager and image analyzer software (AIDA Image Analyzer; Raytest) was used to quantify the radioactively labeled NF-κΒ by densitometry. For comparison between individual gels, the intensity of each band was related to that of simultaneously loaded control animals which had not undergone surgical instrumentation and CLP. Therefore, the EMSA data are expressed as fold increase 25 over control values. Statistics: All data are presented as median (range) unless otherwise stated differences between the two groups were analyzed with the Mann-Whitney rank sum WO 2013/072513 PCT/EP2012/072932 91 test for unpaired samples. Results: The animals treated with NT-M presented with significantly attenuated liver tissue NF-κΒ activation (2.27 (1.97-2.53)) compared to vehicle animals (2.92 (2.50-3.81)) (pO.001) (see figure 10).
References: 1. Wagner F, Wagner K, Weber S, Stahl B, Kndferl MW, Huber-Lang M, Seitz DH, Asfar P, Calzia E, Senftleben U, Gebhard F, Georgieff M, Radermacher P, Hysa V: Inflammatory effects of hypothermia and inhaled H2S during resuscitated, hyperdynamic murine septic shock. Shock 201l;35(4):396-402 2. Wagner F, Scheuerle A, Weber S, Stahl B, McCook O, Knoferl MW, Huber-Lang M, Seitz DH, Thomas J, Asfar P, Szabo C, Moller P, Gebhard F, Georgieff M, Calzia E, Radermacher P, Wagner K: Cardiopulmonary, histologic, and inflammatory effects of intravenous Na2S after blunt chest trauma-induced lung contusion in mice. J Trauma 2011 ;71 (6): 1659-67
Example 6
In vivo side effect determination of antibody NT-M 12-15 week old male C57B1/6 mice (Charles River Laboratories, Germany) were used for the study. 6 mice were treated with (10ul/ g bodyweight) dose of NT-M, 0.2 mg/ml. As control, 6 mice were treated with (ΙΟμΙ/g body weight) PBS. Survival and physical condition was monitored for 14 days. The mortality was 0 in both groups, there were no differences in physical condition between NT-M and control group.
Example 7
Gentamicin-induced nephrotoxicity A non-septic acute kidney injury model has been established, which makes use of the nephrotoxicity induced by Gentamicin (Chiu PJS. Models used to assess renal functions. Drug Develop Res 32:247-255, 1994.), This model was used to assess whether treatment with anti-Adrenomedullin antibody can improve kidney function. WO 2013/072513 PCT/EP2012/072932 92
The experiment was performed as follows:
Effect of a NT-M on Gentamicin-Induced Nephrotoxicity in Rats
Study Design:
Test Cone Dosage Ratsd Group Article Route mg/ml ml/kg mg/kg (Male) 1 Gentamicin3 + vehicle13 IV NA x 4C 8 2 Gentamicin3 + NT-M IV X 4C 8 “Gentamicin at 120 mg/kg intramuscularly for 7 days (days 0-6).
Groups of 8 male Sprague-Dawley rats weighing 250 ± 20 g were employed. Animals were challenged with gentamicin at 120 mg/kg i.m. for seven consecutive days (Groups 1 and 2). 5 Test compound (anti-adrenomedullin antibody NT-M) and vehicle (phosphate buffered saline) were injected intravenously 5 min before gentamicin on day 0, followed by injection on days 2, 4, and 6. Body weights and clinical signs were monitored daily. Twenty-four (24) hour urine collections on ice were performed on Days 0, 2, and 6. Urine specimens were assayed for concentrations of Na+ and K+, and creatinine. Blood samples for clinical 10 chemistry were collected on Days 1 (before gentamicin), 3 (before gentamicin), and 7. Serum electrolytes (Na+ and K+), creatinine, and BUN were the primary analytes that were monitored for assessing renal function. Plasma samples were collected in EDTA tubes (Days 1 and 3:100 μΐ; Day 7:120 μΐ). Creatinine clearance was calculated. Urine volume, urinary WO 2013/072513 PCT/EP2012/072932 93 electrolytes, and creatinine are expressed as amount excreted per 100 g of animal body weight. All animals were sacrificed on Day 7. Kidneys were weighed.
Urine collection. The animals were placed in individual cages where urine was collected for 24 h on Day 0, Day 2, and Day 6. Urine volume, urinary Na+, K+, and creatinine were 5 measured.
Endogenous creatinine clearance was calculated as follows: CCr (ml/24 h) - [UCr (mg/xnl) x V (ml/24 h)] / SCr (mg/ml) 24-hr urinary excretion of sodium (Na+) was calculated as follows: UNaV (pEq/24 h) = UNa (pEq/ml) x V (ml/24 h) 10 24-hr urinary excretion of NAG and NGALwas similarly calculated.
The fractional excretion of Na+ (FE»a)s or percentage of the filtered sodium that is excreted into the final urine, is a measure of tubular Na+reabsorptive function. It was computed as follows: 15 FENa (%) =100 x [UNa (pEq/ml) x V (ml/24 h)] / PNa (pEq/ml) X CCr (ml/24 h)
Treatment with anti-Adrenomedullin antibody improved several measures of kidney function on day 7 as compared to vehicle: serum creatinine 1.01 mg/dL (NT-M) vs 1.55 mg/dL (vehicle) (Fig. 11), BUN 32.08 mg/dL(NT-M) vs. 52.41 mg/dL (vehicle) (Fig. 12), endogenous creatinine clearance 934.43 mL/24 h (NT-M) vs. 613.34 mL/24 h (vehicle) (Fig. 20 13), fractional secretion ofNa+ 0.98 % (NT-M) vs. 1.75 % (vehicle) (Fig. 14).
Example 8
In the mice CLP model described above, the effect of treatment with anti-adrenomedullin antibody NT-M on several parameters of kidney function was investigated. 25 NT-M caused a three- and two-fold higher diuresis and creatinine clearance, respectively, ultimately resulting in lower creatinine, urea, and NGAL blood concentrations at the end of the experiment (see Table 10). Moreover, keratinocyte-derived chemokine (KC) concentrations in the kidney were significantly lowered by treatment with NT-M (Fig. 15). WO 2013/072513 PCT/EP2012/072932 94
Table 10: Parameters of kidney function in the vehicle- (n=l 1) and NT-M-treated (n=9) animals. Blood concentrations were measured in samples taken at the end of the experiment. NGAL = neutrophil gelatinase-associated lipocalin. All data are median (quartiles).
Vehicle NT-M p-Value Urine output [pL-g"1-h'1] 4.4 (3.5;16.5) 15.2 (13.9-,22.5) 0.033 Creatinine clearance [pL-min'] 197 (110;301) 400 (316-,509) 0.006 Creatinine [pg-mL"1 j 1.83 (1.52;3.04) 1.28 (1.20; 1.52) 0.010 Urea [pg-mL'1] 378 (268;513) 175 (101; 184) 0.004 NGAL [pg-mL'1] 16 (15;20) 11 (10;13) 0.008 5
The experiments were performed as follows:
Creatinine, urea, and neutrophil gelatinase-associated lipocalin (NGAL)
Blood NGAL concentrations were measured using a commercial ELISA (mouse NGAL, 10 RUO 042, BioPorto Diagnostics A/S, Denmark, Gentofte). Urea and creatinine concentrations were measured with a capillary column (Optima-5MS, Macherey-Nagel, Diiren, Germany) gas chromatography/mass spectrometry system (Agilent 5890/5970, Boblingen, Germany) using 2H3-creatinine (CDN isotopes, Pointe-Claire, QU, Canada) and methyl-urea (FlukaChemikalien, Buchs, Switzerland) as internal standards. After deproteinization with 15 acetonitrile, centrifugation and evaporation to dryness, the supernatant was reconstituted in formic acid, and extracted over a weak anion exchange column (WCX, Phenomenex, Aschaffenburg, Germany). Acetonitrile plus N,0-Bis(trimethylsilyl)trifluoroacetamide and N-(tert-butyldimethylsilyl)-N-methyltrifluoroacetamide allowed formation of the urea tert-butyl-dimethylsilyl- and the creatininetrimethylsilyl-derivatives, respectively, Ions m/z 231 and 20 245, and m/z 329 and 332 were monitored for urea and creatinine analytes and internal standards, respectively. From the urine output and the plasma and urine creatinine concentrations creatinine clearance was calculated using the standard formula.
Sample preparation WO 2013/072513 PCT/EP2012/072932 95
The kidney which was stored at -80°C was disrupted with a homogenizer in PBS and lysed with a 2-fold concentrated buffer for a whole cell lysate (100 mM Tris pH 7,6; 500 mM NaCl; 6 mM EDTA; 6 mM EGTA; 1 % Triton-X-100; 0,5 % NP 40; 10 % Glycerol; Protease-5 Inhibitors (β-Glycerolphosphate 2 mM; DTT 4 mM; Leupeptine 20 μΜ; Natriumorthovanadate 0,2 mM)) and subsequently centrifuged. The whole cell lysate was obtained out of the supernatant; the pellet consisting of cell remnants was discarded. The amount of protein was determined photometrically with a commercially available protein assay (Bio-Rad, Hercules, CA) and the specimens were adjusted in the way that the final 10 protein concentration was 4 pg/μΐ. The samples for the Multiplex- and EMSA analysis were diluted 1:1 with EMSA buffer (10 mM Hepes; 50 mM KC1; 10 % Glycerol; 0,1 mM EDTA; 1 mM DTT), the samples for the immuno blots 1:1 with 2-fold Sample Buffer (2 % SDS; 125 mM Tris-HCL (pH 6,8 at 25°C); 10 % Glycerol; 50 mM DTT; 0,01 % Bromophenol blue). 15 Levels of keratinocyte-derived chemokine (KC) concentrations were determined using a mouse multiplex cytokine kit (Bio-Plex Pro Cytokine Assay, Bio-Rad, Hercules, CA), the assay was performed by using the Bio-plex suspension array system with the manufacturer’s instructions (see also Wagner F, Wagner K, Weber S, Stahl B, Knoferl MW, Huber-Lang M, Seitz DH, Asfar P, Calzia E, Senftleben U, Gebhard F, Georgieff M, Radermacher P, Hysa V. 20 Inflammatory effects of hypothermia and inhaled H2S during resuscitated, hyperdynamic murine septic shock. Shock 2011;35:396-402; and Wagner F, Scheuerle A, Weber S, Stahl B, McCook O, Knoferl MW, Huber-Lang M, Seitz DH, Thomas J, Asfar P, Szabo C, Moller P, Gebhard F, Georgieff M, Calzia E, Radermacher P, Wagner K. Cardiopulmonary, histologic, and inflammatory effects of intravenous Na2S after blunt chest trauma-induced lung 25 contusion in mice. J Trauma 2011;71:1659-1667). In brief, the appropriate cytokine standards and samples were added to a filter plate. The samples were incubated with antibodies chemically attached to fluorescent-labeled micro beads. Thereafter, premixed detection antibodies were added to each well, and subsequently, streptavidin-phycoeiythdn was added. Beads were then re-suspended, and the cytokines reaction mixture was quantified using the 30 Bio-Plex protein array reader. Data were automatically processed and analyzed by Bio-Plex Manager Software 4.1 using the standard curve produced from recombinant cytokine standards. Levels below the detection limit of the assays were set to zero for statistical purposes.
Example 9 WO 2013/072513 PCT/EP2012/072932 96
In the mice CLP model described above, the effect of treatment with anti-adrenomedullin antibody NT-M on the liver was investigated. NT-M caused a significant lowering of keratinocyte-derived cheraokine (KC) concentrations in the liver (Fig. 16).
Measurement of keratinocyte-derived chemokine (KC) was done analogous to example 8 (kidney).
Example 10
In the mice CLP model described above, the effect of treatment with anti-adrenomedullin antibody NT-M on several cytokines and chemokinesin the blood circulation (plasma) was investigated.
Cytokine and chemokine concentrations
Plasma levels of tumor necrosis factor (TNF)-a, interleukin (IL)-6, monocyte chemoattractant protein (MCP)-l, and keratinocyte-derived chemokine (KC) concentrations were determined using a mouse multiplex cytokine kit (Bio-Plex Pro Cytokine Assay, Bio-Rad, Hercules, CA), the assay was performed by using the Bio-plex suspension array system with the manufacturer’s instructions (see also Wagner F, Wagner K, Weber S, Stahl B, Knoferl MW, Huber-Lang M, Seitz DH, Asfar P, Calzia E, Senftleben U, Gebhard F, Georgieff M, Radermacher P, Hysa V. Inflammatory effects of hypothermia and inhaled H2S during resuscitated, hyperdynamic murine septic shock. Shock 2011;35:396-402; and Wagner F, Scheuerle A, Weber S, Stahl B, McCook O, Knoferl MW, Huber-Lang M, Seitz DH, Thomas J, Asfar P, Szabo C, Moller P, Gebhard F, Georgieff M, Calzia E, Radermacher P, Wagner K. Cardiopulmonary, histologic, and inflammatory effects of intravenous Na2S after blunt chest trauma-induced lung contusion in mice. J Trauma 2011;71:1659-1667). In brief, the appropriate cytokine standards and samples were added to a filter plate. The samples were incubated with antibodies chemically attached to fluorescent-labeled micro beads. Thereafter, premixed detection antibodies were added to each well, and subsequently, streptavidin-phycoerythrin was added. Beads were then re-suspended, and the cytokines reaction mixture was quantified using the Bio-Plex protein array reader. Data were automatically processed and analyzed by Bio-Plex Manager Software 4.1 using the standard curve produced from recombinant cytokine standards. Levels below the detection limit of the assays were set to zero for statistical purposes. WO 2013/072513 PCT/EP2012/072932 97
Plasma levels and kidney tissue concentrations of tumor necrosis factor (TNF)-ot, interleukin (IL)-6 and IL-10, monocyte chemoattractant protein (MCP)-l, and keratinocyte-dervived chemokine (KC) were determined using a commercially available “Multiplex Cytokine Kit” (Bio-Plex Pro Precision Pro Cytokine Assay, Bio-Rad, Hercules, CA), which allows to collect several parameters out of one single sample. The individual work steps of the assay were performed according to the manufacturer's instructions (see also Wagner F, Wagner K, Weber S, Stahl B, Knoferl MW, Huber-Lang M, Seitz DH, Asfar P, Calzia E, Senftleben U, Gebhard F, GeorgiefF M, Radermacher P, Hysa V. Inflammatory effects of hypothermia and inhaled H2S during resuscitated, hyperdynamic murine septic shock. Shock 2011;35:396-402; and Wagner F, Scheuerle A, Weber S, Stahl B, McCook O, Knoferl MW, Huber-Lang M, Seitz DH, Thomas J, Asfar P, Szabo C, Moller P, Gebhard F, Georgieff M, Calzia E, Radermacher P, Wagner K. Cardiopulmonary, histologic, and inflammatory effects of intravenous Ma2S after blunt chest trauma-induced lung contusion in mice. J Trauma 2011;71:1659-1667).
In brief, the fluorescence-labed microspheres (“beads”) were added to a 96-well plate, followed by two washing steps, the addition of internal standards and the addition of plasma-and kidney homogenate samples. During the subsequent incubation the single cytokines bind to the antibodies attached to polystyrene-beads. After the addition of the cytokine-specific biotin-labeled antibodies, which are for the detection of the single cytokines, and an additional incubation time, subsequently phycoerythrin-labeled streptavidine was added. After an additional incubation time, beads were then resuspended, and the plates could be measured with a specific flow cytometer (Bio-Plex suspension array system, Bio-Rad, Hercules, CA). Data were automatically processed and analyzed by Bio-Plex Manager Software 4.1 using the standard curve produced from recombinant cytokine standards. For the plasma levels the concentration was provided in pg * mL'1, the concentration of the kidney homogenates were converted to the appropriate protein concentration and provided in pg * mg'1 protein. NT-M caused a significant lowering of plasma concentrations of IL-6 (Fig. 17), IL-10 (Fig. 18), keratinocyte-derived chemokine (KC) (Fig. 19), monocyte chemoattractant protein-1 (MCP-1) (Fig. 20), TNF-alpha (Fig. 21).
Example 11
Ischemia/Reperfusion-Induced Acute Kidney Injury WO 2013/072513 PCT/EP2012/072932 98
Another non-septic acute kidney injury model has been established, where acute kidney injury is induced by ischemia/reperfusion (Nakamoto M, Shapiro JI, Shanley PF, Chan L, and Schrier RW. In vitro and in vivo protective effect of atriopeptin ill on ischemic acute renal failure. J Clininvest 80:698-705, 1987., Chintala MS, Bernardino V, and Chiu PJS. Cyclic 5 GMP but not cyclic AMP prevents renal platelet accumulation following ischemia-reperfusion in anesthetized rats. J PharmacolExpTher 271:1203-1208, 1994). This model was used to assess whether treatment with anti-adrenomedullin antibody can improve kidney function.
The experiment was performed as follows: 10
Effect of a NT-M on Acute Kidney Injury Induced by Ischemia/Reperfusion in Rats
Study Design:
Test Cone Dosage Rats Group Article Route rne/ml ml/kg mg/kg IMalel 1 I-R + vehicle3 IV 5 NA x 3 8 2 I-R + NT-M IV 5 x 3b 8 a vehicle; injected intravenously (i.v.) 5 min before reperfusion on day 0, followed by injections on days 1 and 2. ^T-M at 4 mg/kg was injected intravenously (i.v.) 5 min before reperfusion on day 0, followed by 2 mg/kg i.v. each on days 1 and 2. °Urine collection on days -1,0, 1 and 2, with blood chemistry and urine analysis on days 0, 1, 2 and 3, respectively. Plasma samples were collected in EDTA tubes (Days 0 (immediate before surgery), 1, 2: 100 μΐ, before vehicle or TA; Day 3:120 μΐ.
Clinical observations: daily before surgery, following surgery and throughout treatment.
Groups of 8 male Sprague-Dawley rats weighing 250 to 280 g were used. The animals were kept on a 12-hr light/dark cycle and receive a standard diet with distilled water ad libitum. The animals receive fluid supplements (0.9% NaCl and 5% dextrose/l:l, 10 ml/kg p.o.) 30 15 min prior to surgery (day 0). The rats were anaesthetized with pentobarbital (50 mg/kg, i.p.). The abdominal cavity was exposed via a midline incision, followed by intravenous administration of heparin (100 U/kg, i.v.) and both renal arteries were occluded for 45 min by WO 2013/072513 PCT/EP2012/072932 99 using vascular clamps. Immediately after removal of the renal clips, the kidneys were observed for additional 1 min to ensure color change indicating blood reperfusion. The test compound (NT-M) and vehicle (phosphate buffered saline) were injected intravenously 5 min before reperfusion, followed by daily injection on days 1 and 2. 5 Urine collection. The 24-h urine collection on ice was initiated at 24h before ischemia/reperfusion on day -1 (-24h to Oh), and day 0 (0-24h), day 1 (24-48h) and day 2 (48-72h) after reperfusion,
Blood collection: 0.4 ml blood was collected through the tail vein into EDTA tubes at Oh (before I RI surgery), 24h (before vehicle or TA), 48h (before vehicle or TA) and 72h for 10 determination of plasma creatinine/Na+/K+, and BUN; 2 ml blood was collected through venal cava terminally.
The animals were placed in individual cages where urine was collected for 24 h day -1 (-24h-Oh), day 0 (0-24h), day 1 (24-48h) and day 2 (48-72h) after reperfusion on day 0. Urine volume, urinary Na+, K+, and creatinine were measured. 15 The creatinine clearance (CCr) was calculated as follows: CCr (ml/24 h) = [UCr (mg/ml) x V (ml/24 h)] / PCr (mg/ml)
The 24-hr urinary excretion of sodium (Na+) was calculated as follows: UNaV (pEq/24 h) = UNa (pEq/ml) x V (ml/24 h)
The fractional excretion of Na+ (FENa), or percentage of the filtered sodium that is excreted 20 into the final urine, is a measure of tubular Na+ reabsorptive function. It was computed as follows: FENa (%) =100 x [UNa (pEq/ml) x V (ml/24 h)] / PNa (pEq/ml) X CCr (ml/24 h)
Treatment with anti-Adrenomedullin antibody improved several measures of kidney function: 25 Blood urea nitrogen (BUN) showed a strong increase in the vehicle group (0 h: 17.49 mg/dL, 24 h; 98.85 mg/dL, 48 h: 109.84 mg/dL, 72 h: 91.88 mg/dL), which was less pronounced with NT-M treatment (0 h: 16.33 mg/dL, 24 h: 84.2 mg/dL, 48 h: 82.61 mg/dL, 72 h: 64.54 mg/dL) (Fig. 22). WO 2013/072513 PCT/EP2012/072932 100
Seram creatinine developed similarity: Vehicle group (0 h: 0.61 mg/dL, 24 h: 3.3 mg/dL, 48 h: 3.16 mg/dL, 72 h: 2.31 mg/dL), NT-M group: (0 h: 0.59 mg/dL, 24 h: 2.96 mg/dL, 48 h: 2.31 mg/dL, 72 h: 1.8 mg/dL) (Fig. 23).
The endogenous creatinine clearance dropped massively on day one and thereafter improved better in the NT-M group than in the vehicle group. Vehicle group: (0 h: 65.17mL/h, 24 h: 3.5mL/h, 48 h: 12.61mL/h, 72 h: 20.88mL/h), NT-M group:(0 h: 70.11mL/h, 24 h: 5.84mL/h, 48 h: 21.23mL/h, 72 h: 26.61mL/h) (Fig. 24). bVehicle; injected intravenously (i.v.) 5 min before gentamicin on Day 0, followed by injections on Days 2, 4, and 6. CNT-M at 4 mg/kg was injected intravenously (i.v.) 5 min before gentamicin on Day 0, followed by 2 mg/kg i.v. on Days 2, 4, and 6. dPlasma samples were collected in EDTA tubes (Days 1 and 3 before Test and Control article: 100 μΐ; Day 7:120 μΐ. 24h urine collection on ice is initiated after gentamicin on Day 0, followed by Days 2 and 6; blood collection on days 1,3, and 7. WO 2013/072513 PCT/EP2012/072932 101
FIGURE DESCRIPTION
Fig. la:
Illustration of antibody formats - Fv and scFv-Variants Fig lb:
Illustration of antibody formats - heterologous fusions and bifunctional antibodies Fig lc:
Illustration of antibody formats - bivalental antibodies and bispecific antibodies Fig. 2: hADM 1-52 (SEQ ID No. 21) mADM 1-50 (SEQ ID No. 22) aa 1-21 of human ADM (SEQ ID No. 23) aa 1-42 of human ADM (SEQ ID No. 24) aa 43-52 of human ADM (SEQ ID No. 25) aa 1-14 of human ADM (SEQ ID NO: 26) aa 1-10 of human ADM (SEQ ID NO: 27) aa 1-6 of human ADM (SEQ ID NO: 28) aa 1-32 of human mature human ADM (SEQ ID NO: 29) aa 1-40 of mature murine ADM (SEQ ID NO: 30) aa 1-31 of mature murine ADM (SEQ ID NO: 31)
Fig. 3: a: Dose response curve of human ADM. Maximal cAMP stimulation was adjusted to 100% activation b: Dose/ inhibition curve of human ADM 22-52 (ADM-receptor antagonist) in the presence of 5.63nM hADM. c: Dose/ inhibition curve of CT-H in the presence of 5.63 nM hADM. d: Dose/ inhibition curve of MR-H in the presence of 5.63 nM hADM. e: Dose/ inhibition curve of NT-H in the presence of 5.63 nM hADM. WO 2013/072513 PCT/EP2012/072932 102 f: Dose response curve of mouse ADM. Maximal cAMP stimulation was adjusted to 100% activation g: Dose/ inhibition curve of human ADM 22-52 (ADM-receptor antagonist) in the presence of 0,67 nMm ADM.
h: Dose/ inhibition curve of CT-M in the presence of 0,67 nM mADM. i: Dose/ inhibition curve of MR-M in the presence of 0,67 nM mADM. j: Dose/ inhibition curve of NT-M in the presence of 0,67 nM mADM. k: shows the inhibition of ADM by F(ab)2 NT-M and by Fab NT-M
1: shows the inhibition of ADM by F(ab)2 NT-M and by Fab NT-M
Fig. 4:
This figure shows a typical hADM dose/ signal curve. And an hADM dose signal curve in the presence of 100 pg/mL antibody NT-H.
Fig. 5:
This figure shows the stability of hADM in human plasma (citrate) in absence and in the presence of NT-H antibody.
Fig. 6:
Alignment of the Fab with homologous human framework sequences Fig. 7:
This figure shows the Noradrenalin requirements for early and late treatment with NT-M Fig. 8:
This figure shows urine production after early and late treatment with NT-M Fig. 9:
This figure shows the fluid balance after early and late treatment with NT-M
Fig. 10:
Liver tissue activation of nuclear factor kappa-light-chain gene enhancer in B cells (NF-kB) analyzed by electophoretic mobility shift assay (EMSA). # depicts p<0.001 vs. vehicle. WO 2013/072513 PCT/EP2012/072932 103
Fig. 11:
Development of serum creatinine over time. Mean +/- SEM are shown.
Fig. 12:
Development of blood urea nitrogen (BUN) over time. Mean +/- SEM are shown. 5 Fig. 13:
Development of endogenous creatinine clearance over time. Mean +/- SEM are shown.
Fig. 14:
Development of fractional secretion of Na+ over time. Mean +/- SEM are shown. 10
Fig. 15:
Keratinocyte-derived chemokine (KC) levels determined in relation to the total kidney protein extracted. The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M. 15 Fig. 16:
Keratinocyte-derived chemokine (KC) levels determined in relation to the total liver protein extracted. The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M. 20 Fig. 17:
Plasma IL-6 levels. The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
Fig. 18:
Plasma IL-10 levels. The white box-plot shows results obtained with vehicle, the grey box-25 plot shows results obtained after treatment with NT-M. I l:\grs\Inta-wovcn\NRPoribl\DCC\GRSVI5174781 I .docx-1107 2017 2012338733 11 Μ 2017 104
Fig. 19:
Plasma keratinocyte-derived chemokine (KC) levels. The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M. 5 Fig. 20:
Plasma monocyte chemoattractant protein-1 (MCP-1) levels. The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
Fig. 21: 10 Plasma TNF-alpha levels. The white box-plot shows results obtained with vehicle, the grey box-plot shows results obtained after treatment with NT-M.
Fig. 22:
Development of blood urea nitrogen (BUN) over time. Mean +/- SEM are shown. 15
Fig. 23:
Development of serum creatinine over time. Mean +/- SEM are shown.
Fig. 24: 20 Development of endogenous creatinine clearance over time. Mean +/- SEM are shown.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but 25 not the exclusion of any other integer or step or group of integers or steps.
The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from 30 it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

Claims (23)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
    1. A method of stabilizing the systemic circulation of a patient in need thereof by preventing or treating an acute disease or acute condition in said patient, the method comprising administering an effective dose of an anti-Adrenomedullin (ADM) antibody, an anti-ADM antibody fragment binding to adrenomedullin or an anti-ADM non-Ig protein scaffold binding to adrenomedullin to said patient, wherein said antibody, antibody fragment or non-Ig scaffold binds to a region of at least 4 amino acids within the sequence of amino acids 1-42 of mature human ADM as set out in SEQ ID NO: 24.
  2. 2. The method of claim 1, wherein a) said patient is in need of stabilizing the systemic circulation and exhibits a heart rate of > 100 beats /min and/or < 65 mm Hg mean arterial pressure and wherein stabilizing the systemic circulation means increasing the mean arterial pressure over 65 mmHg, or b) administering said antibody, antibody fragment or non-Ig scaffold prevents the heart rate increasing to > 100 beats /min and/or mean arterial pressure decreasing to < 65 mm Hg in said patient.
  3. 3. The method of claim 1 or 2, wherein said antibody, antibody fragment or non-Ig scaffold reduces the vasopressor requirement of said patient.
  4. 4. The method of claim 3, wherein the vasopressor comprises catecholamine.
  5. 5. The method of any one of claims 1 to 4, wherein said antibody, antibody fragment or non-Ig scaffold is monospecific.
  6. 6. The method of any one of claims 1 to 5, wherein said antibody, antibody fragment or non-Ig scaffold exhibits a binding affinity to ADM of at least 10'7 M.
  7. 7. The method of any one of claims 1 to 6, wherein said antibody, antibody fragment or non-Ig scaffold is not ADM-binding-Protein-1 (complement factor H).
  8. 8. The method of any one of claims 1 to 7, wherein said antibody, antibody fragment or non-Ig scaffold binds to a region of at least 4 or at least 5 amino acids within the sequence of amino acids 1-21 of mature human ADM: YRQSMNNFQGLRSFGCRFGTC (SEQ ID NO: 23).
  9. 9. The method of any one of claims 1 to 8, wherein said antibody, antibody fragment or non-Ig scaffold recognizes and binds to an epitope containing the N-terminal end amino acid of mature human ADM and wherein said antibody, antibody fragment or non-Ig scaffold does not bind N-terminal extended nor N-terminal modified adrenomedullin or N-terminal degraded adrenomedullin.
  10. 10. The method any one of claims 1 to 9, wherein said antibody, antibody fragment or non-Ig scaffold is an ADM stabilizing antibody, antibody fragment or non-Ig scaffold that enhances the half life (½ half retention time) of adrenomedullin in serum, blood or plasma by at least 10 %, at least 50 %, greater than 50 %, or greater than 100 %.
  11. 11. The method of any one of claims 1 to 10, wherein said antibody, antibody fragment or non-Ig scaffold blocks the circulating ADM bioactivity not more than 80 % or not more than 50%, wherein the bioactivity of ADM is determined in a human recombinant Adrenomedullin receptor cAMP functional assay.
  12. 12. The method of any one of claims 1 to 11, wherein said antibody, antibody fragment or non-Ig scaffold is a modulating anti-ADM antibody, modulating anti-adrenomedullin antibody fragment or modulating anti-ADM non-Ig scaffold that enhances the half life (½ half retention time) of adrenomedullin in serum, blood or plasma by at least 10 %, at least 50 %, greater than 50 %, or greater than 100 % and that blocks the bioactivity of ADM to not more than 80 % or not more than 50 %, wherein the bioactivity of ADM is determined in a human recombinant Adrenomedullin receptor cAMP functional assay.
  13. 13. The method of any one of claims 1 to 12, wherein said antibody or antibody fragment is a human monoclonal antibody or antibody fragment that binds to ADM or a fragment thereof, wherein the heavy chain of said antibody or antibody fragment comprises the sequences SEQ ID NO: 1 GYTFSRYW SEQ ID NO: 2 ILPGSGST SEQ ID NO: 3 TEGYEYDGFDY and wherein the light chain comprises the sequences SEQ ID NO:4 QSIVYSNGNTY SEQ ID NO: 5 RVS SEQ ID NO: 6 FQGSHIPYT.
  14. 14. The method of claim 13, wherein said antibody or antibody fragment comprises at least one of the sequences: SEQ ID NO: 7 (AM-VH-C) QVQLQQSGAELMKPGASVKISCKATGYTFSRYWIEWVKQRPGHGLEWIGEIL PGSGSTNYNEKFKGKATITADTSSNTAYMQLSSLTSEDSAVYYCTEGYEYDGF DYWGQGTTLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVS WNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTK VDKRVEPKHHHHHH SEQ ID NO: 8 (AM-VHI) Q V QLVQS GAE VKKPGS S VKV SCKASG YTFSRY WIS WVRQAPGQGLE WMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 9 (AM-VH2-E40) QV QLVQSGAEVKKPGS S VKVSCKASGYTFSRYWIEWVRQAPGQGLEWMGRI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHH SEQ ID NO: 10 (AM-VH3-T26-E55) QVQLVQSGAEVKKPGSSVKVSCKATGYTFSRYWISWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHIHHHH SEQ ID NO: 11 (AM-VH4-T26-E40-E55) QV QLV QSGAE VKKPGS S VKV SCKATGYTFSRYWIEWVRQAPGQGLEWMGEI LPGSGSTNYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTEGYEYDG FDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWN SGALTSGVHTFPAVLQSSGLYSLSSWTVPSSSLGTQTYICNVNHKPSNT KVDKRVEPKHHHHHH SEQ ID NO: 12 (AM-VL-C) DVLLSQTPLSLPVSLGDQATISCRSSQSrVYSNGNTYLEWYLQKPGQSPKLLIY RV SNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYY CF QGSHIP YTF GGGTK LEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQS GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSF NRGEC SEQ ID NO: 13 (AM-VL1) DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLNWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQ SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC or SEQ ID NO: 14 (AM-VL2-E40) DVVMTQSPLSLPVTLGQPASISCRSSQSIVYSNGNTYLEWFQQRPGQSPRRLIY RVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTFGQGT KLEIKRTVAAPSVFIFPPSDEQLKSGTASWCLLNNFYPREAKVQWKVDNALQ SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKS FNRGEC.
  15. 15. The method of any one of claims 1 to 14, wherein said patient suffers from one or more of SIRS, sepsis, diabetes, cancer, acute vascular disease, shock or organ dysfunction.
  16. 16. The method of claim 15, wherein acute vascular disease comprises heart failure, shock comprises septic shock and organ dysfunction comprises kidney dysfunction.
  17. 17. The method of any one of claims 1 to 16, wherein said antibody, antibody fragment or non-Ig scaffold does not bind to the C-terminal portion of ADM, amino acids 43-52 of ADM: PRSKISPQG Y -NH2 (SEQ ID NO: 25).
  18. 18. The method of any one of claims 1 to 17 , further comprising administration of catecholamine and/or one or more fluids, wherein the catecholamine and/or fluids are administered intravenously.
  19. 19. The method of any one of claims 1 to 18, further comprising administration of ADM binding protein and/or one or more further active ingredients.
  20. 20. Use of an anti-Adrenomedullin (ADM) antibody, anti-ADM antibody fragment binding to adrenomedullin or anti-ADM non-Ig protein scaffold binding to adrenomedullin in the manufacture of a medicament for preventing or treating an acute disease or acute condition in a patient in need thereof, wherein said antibody, antibody fragment or non-Ig scaffold binds to a region of at least 4 amino acids within the sequence of amino acids 1-42 of mature human ADM as set out in SEQ ID NO: 24.
  21. 21. Use of claim 20, wherein said medicament is formulated as a solution or a freeze-dried state.
  22. 22. Use of claim 21, wherein said solution is a ready-to-use solution.
  23. 23. Use of any one of claims 21 to 23, wherein said medicament is for stabilizing the systemic circulation of the patient.
AU2012338733A 2011-11-16 2012-11-16 Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation Active AU2012338733B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP11189449.9 2011-11-16
EP11189449 2011-11-16
EP12160016 2012-03-16
EP12160016.7 2012-03-16
PCT/EP2012/072932 WO2013072513A1 (en) 2011-11-16 2012-11-16 Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation

Publications (2)

Publication Number Publication Date
AU2012338733A1 AU2012338733A1 (en) 2014-05-29
AU2012338733B2 true AU2012338733B2 (en) 2017-08-24

Family

ID=48429016

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012338733A Active AU2012338733B2 (en) 2011-11-16 2012-11-16 Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation

Country Status (15)

Country Link
US (1) US9402900B2 (en)
EP (1) EP2780370B1 (en)
JP (1) JP6224608B2 (en)
AU (1) AU2012338733B2 (en)
CA (1) CA2856142A1 (en)
DK (1) DK2780370T3 (en)
ES (1) ES2751492T3 (en)
HR (1) HRP20191771T1 (en)
HU (1) HUE045940T2 (en)
LT (1) LT2780370T (en)
PL (1) PL2780370T3 (en)
PT (1) PT2780370T (en)
SG (1) SG11201402358RA (en)
WO (1) WO2013072513A1 (en)
ZA (1) ZA201403549B (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3082858T3 (en) * 2013-12-20 2021-09-27 Angiobiomed Gmbh Adrenomedullin binder for use in therapy of cancer
US11076813B2 (en) * 2016-07-22 2021-08-03 Edwards Lifesciences Corporation Mean arterial pressure (MAP) derived prediction of future hypotension
AU2017375049A1 (en) 2016-12-16 2019-06-13 Adrenomed Ag Anti-Adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
EP3339324A1 (en) 2016-12-22 2018-06-27 sphingotec GmbH Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in intervention and therapy of congestion in a patient in need thereof
CA3076691A1 (en) 2017-09-25 2019-03-28 Adrenomed Ag Anti-adrenomedullin (adm) binder for use in therapy or prevention of symptoms of illness
JP2021500548A (en) 2017-10-18 2021-01-07 アドレノメト アクチェンゲゼルシャフト Treatment monitoring under treatment with anti-adrenomedullin (ADM) binder
EP3871689A1 (en) 2020-02-26 2021-09-01 sphingotec GmbH Anti-adm-antibodies binding to the free n-terminus for accelerated transition of adm-gly to bio-adm in patients with adm-gly/ bio-adm ratio above a threshold and combination with vitamin c
MX2022010207A (en) 2020-02-27 2022-11-16 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock.
IL295951A (en) 2020-02-27 2022-10-01 Adrenomed Ag Anti-adrenomedullin (adm) binder for use in therapy of patients in shock
WO2021170838A1 (en) 2020-02-27 2021-09-02 4TEEN4 Pharmaceuticals GmbH Dpp3 for therapy guidance, monitoring and stratification of nt-adm antibodies in patients with shock
WO2021185784A1 (en) 2020-03-16 2021-09-23 Sphingotec Gmbh Pro-adrenomedullin or fragment thereof in patients infected with corona virus and treatments with binder against adrenomedullin
WO2023175035A1 (en) 2022-03-15 2023-09-21 Adrenomed Ag Stable aqueous formulation of an anti-adrenomedullin (adm) antibody or anti-adm antibody fragment
WO2024023369A1 (en) 2022-07-29 2024-02-01 Adrenomed Ag Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for use in therapy or prevention of shock
WO2024023368A1 (en) 2022-07-29 2024-02-01 4TEEN4 Pharmaceuticals GmbH Prediction of an increase of dpp3 in a patient with septic shock
EP4345109A1 (en) 2022-09-30 2024-04-03 AdrenoMed AG Anti-adrenomedullin (adm) binder for use in therapy of pediatric patients with congenital heart disease

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0622458A2 (en) * 1993-04-26 1994-11-02 Shionogi & Co., Ltd. Adrenomedullin

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
AU634716B2 (en) 1988-08-01 1993-03-04 Ciba Corning Diagnostics Corp. Method for detection of an analyte using acridinium esters and liposomes
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DK0463151T3 (en) 1990-01-12 1996-07-01 Cell Genesys Inc Generation of xenogenic antibodies
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
ES2139598T3 (en) 1990-07-10 2000-02-16 Medical Res Council PROCEDURES FOR THE PRODUCTION OF SPECIFIC UNION COUPLE MEMBERS.
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
EP0746609A4 (en) 1991-12-17 1997-12-17 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
CA2229741C (en) 1995-08-18 2012-05-08 The Government Of The United States Of America Functional role of adrenomedullin (am) and the gene-related product (pamp) in human pathology and physiology
CA2242308A1 (en) 1997-12-08 1999-06-08 Smithkline Beecham Laboratoires Pharmaceutiques Novel compounds
DE19847690A1 (en) 1998-10-15 2000-04-20 Brahms Diagnostica Gmbh Diagnosing sepsis and severe infections, useful for assessing severity and progress of treatment, by measuring content of peptide prohormone or their derived fragments
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
AU774725B2 (en) 1999-09-10 2004-07-08 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Determination of adrenomedullin-binding proteins
US7361473B2 (en) 2001-05-04 2008-04-22 Biosite, Incorporated Diagnostic markers of acute coronary syndrome and methods of use thereof
JP2005508623A (en) 2001-08-30 2005-04-07 バイオレクシス ファーマシューティカル コーポレイション Modified transferrin fusion protein
US6864237B2 (en) 2002-05-17 2005-03-08 Ping Wang Treatment of shock using adrenomedullin and adrenomedullin binding protein-1
ES2559927T3 (en) 2002-06-07 2016-02-16 Dyax Corp. Prevention and reduction of blood loss and inflammatory response
DE10316583A1 (en) * 2003-04-10 2004-10-28 B.R.A.H.M.S Aktiengesellschaft Determination of a mid-regional proadrenomedullin partial peptide in biological fluids for diagnostic purposes, as well as immunoassays for carrying out such a determination
JP2007523844A (en) 2003-04-25 2007-08-23 ジェノバ・リミテッド Secreted polypeptide species are reduced in cardiovascular disorders
US6884781B2 (en) 2003-05-16 2005-04-26 Ping Wang Treatment of shock using adrenomedullin binding protein-1
EP1675878A2 (en) 2003-10-24 2006-07-05 Avidia, Inc. Ldl receptor class a and egf domain monomers and multimers
US20100028995A1 (en) 2004-02-23 2010-02-04 Anaphore, Inc. Tetranectin Trimerizing Polypeptides
WO2006027147A2 (en) 2004-09-09 2006-03-16 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with adrenomedullin receptor (amdr)
EP1793847A2 (en) 2004-09-21 2007-06-13 NascaCell IP GmbH Use of microproteins as tryptase inhibitors
JP4976412B2 (en) 2005-12-01 2012-07-18 ベー・エル・アー・ハー・エム・エス・ゲーエムベーハー Methods for diagnosis and treatment of critically ill patients with endothelin, endothelin agonists and adrenomedullin antagonists
MX2010004374A (en) * 2007-10-31 2010-04-30 Medimmune Llc Protein scaffolds.
WO2009077175A1 (en) 2007-12-19 2009-06-25 Affibody Ab Polypeptide derived from protein a and able to bind pdgf
ES2836948T3 (en) 2008-11-03 2021-06-28 Molecular Partners Ag Binding proteins that inhibit VEGF-A receptor interaction
NZ598351A (en) 2009-08-27 2014-08-29 Covagen Ag Il-17 binding compounds and medical uses thereof
CN102753569A (en) 2009-12-14 2012-10-24 塞尔蛋白质股份有限公司 Modified ubiquitin proteins having a specific binding activity for the extradomain b of fibronectin
DE102010040035A1 (en) 2010-03-04 2011-09-08 Robert Bosch Gmbh Improvements to backward analysis to determine error masking factors
CN105949301B (en) 2010-06-08 2020-03-27 皮里斯制药有限公司 Tear lipocalin muteins binding to IL-4 receptor α
FR2964103B1 (en) 2010-08-30 2018-11-23 Universite D'aix-Marseille ANTIBODIES BINDING TO ADRENOMEDULLINE AND RECEPTORS OF ADRENOMEDULLINE AND THEIR USES AS A MEDICINAL PRODUCT
MY174877A (en) 2011-11-16 2020-05-20 Adrenomed Ag Anti-adrenomedullin (adm) antibpdy or anti-adm antibody fragment of anti-adm non-ig scaffold for prevention or reduction of organ dysfunction or organ failure in a patient having a chronic of acute disease or acute condition

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0622458A2 (en) * 1993-04-26 1994-11-02 Shionogi & Co., Ltd. Adrenomedullin

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HYVELIN, J.M. et al. "Adrenomedullin: a cardiac depressant factor in septic shock." J Card Surg. 2002 Jul-Aug;17(4):328-35. *
MARTINEZ, A. et al. "Is adrenomedullin a causal agent in some cases of type 2 diabetes?." Peptides. 1999 Dec;20(12):1471-8. *
OUAFIK, L. et al. "Neutralization of adrenomedullin inhibits the growth of human glioblastoma cell lines in vitro and suppresses tumor xenograft growth in vivo." Am J Pathol. 2002 Apr;160(4):1279-92. *
WANG, P. et al. "The pivotal role of adrenomedullin in producing hyperdynamic circulation during the early stage of sepsis." Arch Surg. 1998 Dec;133(12):1298-304. *

Also Published As

Publication number Publication date
DK2780370T3 (en) 2019-10-28
HRP20191771T1 (en) 2019-12-27
WO2013072513A1 (en) 2013-05-23
JP2015502349A (en) 2015-01-22
LT2780370T (en) 2019-11-25
EP2780370A1 (en) 2014-09-24
ES2751492T3 (en) 2020-03-31
NZ624876A (en) 2016-08-26
CA2856142A1 (en) 2013-05-23
ZA201403549B (en) 2022-11-30
HUE045940T2 (en) 2020-01-28
JP6224608B2 (en) 2017-11-01
PT2780370T (en) 2019-10-30
US20140322225A1 (en) 2014-10-30
EP2780370B1 (en) 2019-09-25
SG11201402358RA (en) 2014-06-27
US9402900B2 (en) 2016-08-02
PL2780370T3 (en) 2020-01-31
AU2012338733A1 (en) 2014-05-29

Similar Documents

Publication Publication Date Title
AU2012338733B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for use in therapy of an acute disease or acute condition of a patient for stabilizing the circulation
AU2012338732B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or an anti-ADM non-Ig scaffold for use in therapy
US11673949B2 (en) Method of modulating the adrenomedullin (ADM) activity of a patient by administering to the patient an anti-ADM antibody or fragment thereof that specifically binds to mature human ADM
AU2012338730B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for reducing the risk of mortality in a patient having a chronic or acute disease or acute condition
AU2012338734B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment or anti-ADM non-Ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease
US9304127B2 (en) Anti-adrenomedullin (ADM) antibody or anti-ADM antibody fragment for use in therapy
CA2856150A1 (en) Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for regulating the fluid balance in a patient having a chronic or acute disease
CA2856154A1 (en) Anti-adrenomedullin (adm) antibody or anti-adm antibody fragment or anti-adm non-ig scaffold for prevention or reduction of organ dysfunction or organ failure in a patient having a chronic or acute disease or acute condition

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)